Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 210(3): 283-296, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36548461

RESUMO

Foot-and-mouth disease virus (FMDV) is the causative agent of foot-and-mouth disease, one of the most highly infectious animal viruses throughout the world. The JAK-STAT signaling pathway is a highly conserved pathway for IFN-ß-induced antiviral gene expression. Previous studies have shown that FMDV can strongly suppress the innate immune response. Moreover, although STAT1 and STAT2 (STAT1/2) have been well established in JAK-STAT signaling-induced antiviral gene expression, whether FMDV proteins inhibit IFN-ß-induced JAK-STAT signaling remains poorly understood. In this study, we described the Lb leader protease (Lbpro) of FMDV as a candidate for inhibiting IFN-ß-induced signaling transduction via directly interacting with STAT1/2. We further showed that Lbpro colocalized with STAT1/2 to inhibit their nuclear translocation. Importantly, Lbpro cleaved STAT1/2 to inhibit IFN-ß-induced signal transduction, whereas the catalytically inactive mutant of LC51A (Lbpro with cysteine substituted with alanine at amino acid residue 51) had no effect on the stability of STAT1/2 proteins. The cleavage of the STAT1/2 proteins was also determined during FMDV infection in vitro. Lbpro could cleave the residues between 252 and 502 aa for STAT1 and the site spanning residues 140 - 150 aa (QQHEIESRIL) for STAT2. The in vivo results showed that Lbpro can cleave STAT1/2 in pigs. Overall, our findings suggest that FMDV Lbpro-mediated targeting of STAT1/2 may reveal a novel mechanism for viral immune evasion.


Assuntos
Endopeptidases , Vírus da Febre Aftosa , Interferon beta , Fator de Transcrição STAT1 , Fator de Transcrição STAT2 , Animais , Vírus da Febre Aftosa/enzimologia , Imunidade Inata , Peptídeo Hidrolases , Transdução de Sinais , Suínos , Interferon beta/imunologia
2.
Wiley Interdiscip Rev RNA ; 12(4): e1645, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33605051

RESUMO

RNA viruses have developed specialized mechanisms to subvert host RNA-binding proteins (RBPs) favoring their own gene expression. The Leader (L) protein of foot-and-mouth disease virus, a member of the Picornaviridae family, is a papain-like cysteine protease that self-cleaves from the polyprotein. Early in infection, the L protease cleaves the translation initiation factors eIF4GI and eIF4GII, inducing the shutdown of cap-dependent translation. However, the cleavage sites on the viral polyprotein, eIF4GI, and eIF4GII differ in sequence, challenging the definition of a consensus site for L targets. Identification of Gemin5 and Daxx proteolytic products in infected cells unveiled a motif centered on the RKAR sequence. The RBP Gemin5 is a member of the survival of motor neurons complex, a ribosome interacting protein, and a translation downregulator. Likewise, the Fas-ligand Daxx is a multifunctional adaptor that plays key roles in transcription control, apoptosis, and innate immune antiviral response. Remarkably, the cleavage site on the RNA helicases MDA5 and LGP2, two relevant immune sensors of the retinoic acid-inducible gene-I (RIG-I)-like receptors family, resembles the L target site of Gemin5 and Daxx, and similar cleavage sites have been reported in ISG15 and TBK1, two proteins involved in type I interferon response and signaling pathway, respectively. In this review we dissect the features of the L cleavage sites in essential RBPs, eventually helping in the discovery of novel L targets. This article is categorized under: RNA in Disease and Development > RNA in Disease Translation > Translation Regulation.


Assuntos
Fatores de Restrição Antivirais/imunologia , Vírus da Febre Aftosa , Imunidade Inata , RNA , Animais , Vírus da Febre Aftosa/enzimologia , Vírus da Febre Aftosa/genética , RNA Helicases
3.
J Virol ; 95(1)2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33028719

RESUMO

The low fidelity of foot-and-mouth disease virus (FMDV) RNA-dependent RNA polymerase allows FMDV to exhibit high genetic diversity. Previously, we showed that the genetic diversity of FMDV plays an important role in virulence in suckling mice. Here, we mutated the amino acid residue Phe257, located in the finger domain of FMDV polymerase and conserved across FMDV serotypes, to a cysteine (F257C) to study the relationship between viral genetic diversity, virulence, and transmissibility in natural hosts. The single amino acid substitution in FMDV polymerase resulted in a high-fidelity virus variant, rF257C, with growth kinetics indistinguishable from those of wild-type (WT) virus in cell culture, but it displayed smaller plaques and impaired fitness in direct competition assays. Furthermore, we found that rF257C was attenuated in vivo in both suckling mice and pigs (one of its natural hosts). Importantly, contact exposure experiments showed that the rF257C virus exhibited reduced transmissibility compared to that of wild-type FMDV in the porcine model. This study provides evidence that FMDV genetic diversity is important for viral virulence and transmissibility in susceptible animals. Given that type O FMDV exhibits the highest genetic diversity among all seven serotypes of FMDV, we propose that the lower polymerase fidelity of the type O FMDV could contribute to its dominance worldwide.IMPORTANCE Among the seven serotypes of FMDV, serotype O FMDV have the broadest distribution worldwide, which could be due to their high virulence and transmissibility induced by high genetic diversity. In this paper, we generated a single amino acid substitution FMDV variant with a high-fidelity polymerase associated with viral fitness, virulence, and transmissibility in a natural host. The results highlight that maintenance of viral population diversity is essential for interhost viral spread. This study provides evidence that higher genetic diversity of type O FMDV could increase both virulence and transmissibility, thus leading to their dominance in the global epidemic.


Assuntos
Vírus da Febre Aftosa/patogenicidade , Febre Aftosa/virologia , RNA Polimerase Dependente de RNA/fisiologia , Proteínas não Estruturais Virais/fisiologia , Animais , Linhagem Celular , Cricetinae , Vírus da Febre Aftosa/enzimologia , Vírus da Febre Aftosa/genética , Aptidão Genética , Variação Genética , Camundongos , Mutação , Fenótipo , RNA Polimerase Dependente de RNA/genética , Suínos , Proteínas não Estruturais Virais/genética , Virulência
4.
Molecules ; 25(16)2020 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-32784761

RESUMO

The development and evaluation of a Boc-AL(Boc)Q(Trt)-AMC fluorophore to detect 3C Protease, produced by Foot and Mouth Disease Virus (FMDV) is reported, with a view to a potential use as a rapid screen for FMDV infected livestock The peptide-linked conjugate fluorophore is evaluated in vitro for sensitivity, specificity, stability and rapidity and shows statistically significant increases in fluorescence when exposed to physiologically relevant concentrations of 3C Protease and selectivity when compared with other common proteases likely to be located, typically in the absence of FMDV. The stability of deprotected Boc-AL(Boc)Q(Trt)-AMC is reported as a limitation of this probe.


Assuntos
Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Ensaios Enzimáticos/métodos , Corantes Fluorescentes/química , Corantes Fluorescentes/síntese química , Vírus da Febre Aftosa/enzimologia , Proteínas Virais/química , Proteínas Virais/metabolismo , Proteases Virais 3C , Linhagem Celular , Técnicas de Química Sintética
5.
J Gen Virol ; 100(3): 446-456, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30702422

RESUMO

The production of experimental molecular vaccines against foot-and-mouth disease virus utilizes the viral encoded 3C protease for processing of the P1 polyprotein. Expression of wild type 3C protease is detrimental to host cells. The molecular vaccine constructs containing the 3C protease L127P mutant significantly reduce adverse effects associated with protease expression while retaining the ability to process and assemble virus-like particles. In published 3C protease crystal structures, the L127 residue is contained within the B2 ß-strand as part of the A2-B2 ß-sheet. To provide insight into the mechanism by which the L127P mutant alters the properties of the 3C protease, we performed scanning proline mutagenesis of residues 123-128 of the B2 ß-strand and monitored expression and P1 processing. Simultaneously, we utilized random mutagenesis of the full 3C sequence to identify additional mutations presenting a phenotype similar to the L127P mutation. Six of the tested mutants enhanced expression over wild type, and the I22P, T100P and V124P mutations surpassed the L127P mutation in certain cell lines. These data areinterpreted in conjunction with published 3C protease crystal structures to provide insight into the mechanism by which these mutations enhance expression.


Assuntos
Cisteína Endopeptidases/química , Cisteína Endopeptidases/genética , Vírus da Febre Aftosa/enzimologia , Febre Aftosa/virologia , Peptídeos/genética , Proteínas Virais/química , Proteínas Virais/genética , Proteases Virais 3C , Animais , Cisteína Endopeptidases/metabolismo , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/metabolismo , Regulação Viral da Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Mutagênese , Peptídeos/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo , Prolina/genética , Prolina/metabolismo , Conformação Proteica em Folha beta , Processamento Pós-Transcricional do RNA , Proteínas Virais/metabolismo
6.
Virology ; 522: 260-270, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30055516

RESUMO

The foot-and-mouth disease virus capsid precursor, P1-2A, is cleaved by the 3C protease (3Cpro) to VP0, VP3, VP1 and 2A. The P1-2A precursor (wt or mutant) was expressed alone or with 3Cpro and processing of P1-2A was determined. The VP2 K217R and VP3 I2P substitutions (near the VP0/VP3 junction) strongly reduced the processing at this junction by 3Cpro while the substitution VP2 K217E blocked cleavage. At the VP3/VP1 junction, the substitutions VP3 Q2221P and VP1 T1P each severely inhibited processing at this site. Blocking cleavage at either junction did not prevent processing elsewhere in P1-2A. These modifications were also introduced into full-length FMDV RNA; only wt and the VP2 K217R mutant were viable. Uncleaved VP0-VP3 and the processed products were observed within cells infected with the mutant virus. The VP0-VP3 was not incorporated into empty capsids or virus particles. The three junctions within P1-2A are processed by 3Cpro independently.


Assuntos
Proteínas do Capsídeo/metabolismo , Cisteína Endopeptidases/metabolismo , Vírus da Febre Aftosa/enzimologia , Vírus da Febre Aftosa/fisiologia , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Virais/metabolismo , Proteases Virais 3C , Animais , Proteólise
7.
PLoS Pathog ; 14(6): e1007135, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29958302

RESUMO

The RNA helicase LGP2 (Laboratory of Genetics and Physiology 2) is a non-signaling member of the retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), whose pivotal role on innate immune responses against RNA viruses is being increasingly uncovered. LGP2 is known to work in synergy with melanoma differentiation-associated gene 5 (MDA5) to promote the antiviral response induced by picornavirus infection. Here, we describe the activity of the foot-and-mouth disease virus (FMDV) Leader protease (Lpro) targeting LGP2 for cleavage. When LGP2 and Lpro were co-expressed, cleavage products were observed in an Lpro dose-dependent manner while co-expression with a catalytically inactive Lpro mutant had no effect on LGP2 levels or pattern. We further show that Lpro localizes and immunoprecipitates with LGP2 in transfected cells supporting their interaction within the cytoplasm. Evidence of LGP2 proteolysis was also detected during FMDV infection. Moreover, the inhibitory effect of LGP2 overexpression on FMDV growth observed was reverted when Lpro was co-expressed, concomitant with lower levels of IFN-ß mRNA and antiviral activity in those cells. The Lpro target site in LGP2 was identified as an RGRAR sequence in a conserved helicase motif whose replacement to EGEAE abrogated LGP2 cleavage by Lpro. Taken together, these data suggest that LGP2 cleavage by the Leader protease of aphthoviruses may represent a novel antagonistic mechanism for immune evasion.


Assuntos
Endopeptidases/metabolismo , Vírus da Febre Aftosa/imunologia , Febre Aftosa/virologia , Evasão da Resposta Imune/imunologia , Imunidade Inata/imunologia , RNA Helicases/metabolismo , Animais , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Endopeptidases/genética , Febre Aftosa/imunologia , Febre Aftosa/patologia , Vírus da Febre Aftosa/enzimologia , Células HEK293 , Humanos , RNA Helicases/genética , RNA Helicases/imunologia , Células Vero
8.
Virology ; 509: 222-231, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28662438

RESUMO

The interferon-induced double-strand RNA activated protein kinase (PKR) plays important roles in host defense against viral infection. Here we demonstrate the significant antiviral role of PKR against foot-and-mouth disease virus (FMDV) and report that FMDV infection inhibits PKR expression and activation in porcine kidney (PK-15) cells. The viral nonstructural protein 3C proteinase (3Cpro) is identified to be responsible for this inhibition. However, it is independent of the well-known proteinase activity of 3Cpro or 3Cpro-induced shutoff of host protein synthesis. We show that 3Cpro induces PKR degradation by lysosomal pathway and no interaction is determined between 3Cpro and PKR. Together, our results indicate that PKR acts an important antiviral factor during FMDV infection, and FMDV has evolved a strategy to overcome PKR-mediated antiviral role by downregulation of PKR protein.


Assuntos
Cisteína Endopeptidases/metabolismo , Vírus da Febre Aftosa/patogenicidade , Interações Hospedeiro-Patógeno , Lisossomos/metabolismo , Proteólise , Proteínas Virais/metabolismo , Replicação Viral , eIF-2 Quinase/antagonistas & inibidores , Proteases Virais 3C , Animais , Linhagem Celular , Vírus da Febre Aftosa/enzimologia , Suínos
9.
J Gen Virol ; 98(4): 671-680, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28452293

RESUMO

Foot-and-mouth disease virus is a picornavirus and its RNA genome encodes a large polyprotein. The N-terminal part of this polyprotein is the leader protein, a cysteine protease, termed Lpro. The virus causes the rapid inhibition of host cell cap-dependent protein synthesis within infected cells. This results from the Lpro-dependent cleavage of the cellular translation initiation factor eIF4G. Lpro also releases itself from the virus capsid precursor by cleaving the L/P1 junction. Using site-directed mutagenesis of the Lpro coding sequence, we have investigated the role of 51 separate amino acid residues in the functions of this protein. These selected residues either are highly conserved or are charged and exposed on the protein surface. Using transient expression assays, within BHK-21 cells, it was found that residues around the active site (W52, L53 and A149) of Lpro and others located elsewhere (K38, K39, R44, H138 and W159) are involved in the induction of eIF4G cleavage but not in the processing of the L/P1 junction. Modified viruses, encoding such amino acid substitutions within Lpro, can replicate in BHK-21 cells but did not grow well in primary bovine thyroid cells. This study characterizes mutant viruses that are deficient in blocking host cell responses to infection (e.g. interferon induction) and can assist in the rational design of antiviral agents targeting this process and in the production of attenuated viruses.


Assuntos
Endopeptidases/metabolismo , Fator de Iniciação Eucariótico 4G/metabolismo , Vírus da Febre Aftosa/enzimologia , Vírus da Febre Aftosa/fisiologia , Proteínas Mutantes/metabolismo , Proteólise , Animais , Bovinos , Células Cultivadas , Cricetinae , Análise Mutacional de DNA , Endopeptidases/genética , Vírus da Febre Aftosa/genética , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética
10.
J Biol Chem ; 290(46): 27618-32, 2015 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-26363073

RESUMO

TRAF family member-associated NF-κB activator (TANK) is a negative regulator of canonical NF-κB signaling in the Toll-like receptor- and B-cell receptor-mediated signaling pathways. However, functions of TANK in viral infection-mediated NF-κB activation remain unclear. Here, we reported that TANK was cleaved by encephalomyocarditis virus 3C at the 197 and 291 glutamine residues, which depends on its cysteine protease activity. In addition, encephalomyocarditis virus 3C impaired the ability of TANK to inhibit TRAF6-mediated NF-κB signaling. Interestingly, we found that several viral proteases encoded by the foot and mouth disease virus, porcine reproductive and respiratory syndrome virus, and equine arteritis virus also cleaved TANK. Our results suggest that TANK is a novel target of some viral proteases, indicating that some positive RNA viruses have evolved to utilize their major proteases to regulate NF-κB activation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cisteína Endopeptidases/metabolismo , Vírus da Encefalomiocardite/enzimologia , NF-kappa B/metabolismo , Proteólise , Fator 6 Associado a Receptor de TNF/metabolismo , Proteínas Virais/metabolismo , Proteases Virais 3C , Sequência de Aminoácidos , Cisteína Endopeptidases/genética , Equartevirus/enzimologia , Vírus da Febre Aftosa/enzimologia , Células HEK293 , Humanos , Dados de Sequência Molecular , Vírus da Síndrome Respiratória e Reprodutiva Suína/enzimologia , Fator 6 Associado a Receptor de TNF/antagonistas & inibidores , Proteínas Virais/genética
11.
Virology ; 468-470: 397-408, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25240326

RESUMO

Translation of foot-and-mouth disease virus RNA initiates at one of two start codons leading to the synthesis of two forms of leader proteinase L(pro) (Lab(pro) and Lb(pro)). These forms free themselves from the viral polyprotein by intra- and intermolecular self-processing and subsequently cleave the cellular eukaryotic initiation factor (eIF) 4 G. During infection, Lb(pro) removes six residues from its own C-terminus, generating sLb(pro). We present the structure of sLb(pro) bound to the inhibitor E64-R-P-NH2, illustrating how sLb(pro) can cleave between Lys/Gly and Gly/Arg pairs. In intermolecular cleavage on polyprotein substrates, Lb(pro) was unaffected by P1 or P1' substitutions and processed a substrate containing nine eIF4GI cleavage site residues whereas sLb(pro) failed to cleave the eIF4GI containing substrate and cleaved appreciably more slowly on mutated substrates. Introduction of 70 eIF4GI residues bearing the Lb(pro) binding site restored cleavage. These data imply that Lb(pro) and sLb(pro) may have different functions in infected cells.


Assuntos
Endopeptidases/metabolismo , Vírus da Febre Aftosa/enzimologia , Sítios de Ligação , Catepsina B/antagonistas & inibidores , Catepsina B/metabolismo , Endopeptidases/genética , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Viral da Expressão Gênica/fisiologia , Modelos Moleculares , Conformação Proteica , RNA Viral
12.
Bioorg Med Chem Lett ; 24(2): 490-4, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24374278

RESUMO

Foot-and-mouth disease virus (FMDV) causes a highly infectious and economically devastating disease of livestock. The FMDV genome is translated as a single polypeptide precursor that is cleaved into functional proteins predominantly by the highly conserved viral 3C protease, making this enzyme an attractive target for antiviral drugs. A peptide corresponding to an optimal substrate has been modified at the C-terminus, by the addition of a warhead, to produce irreversible inhibitors that react as Michael acceptors with the enzyme active site. Further investigation highlighted key structural determinants for inhibition, with a positively charged P2 being particularly important for potency.


Assuntos
Antivirais/síntese química , Antivirais/farmacologia , Cisteína Endopeptidases/química , Desenho de Fármacos , Vírus da Febre Aftosa/efeitos dos fármacos , Vírus da Febre Aftosa/enzimologia , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/química , Proteases Virais 3C , Animais , Cisteína Endopeptidases/metabolismo , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Virais/metabolismo
13.
J Mol Biol ; 425(22): 4530-43, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23973886

RESUMO

EV71 is responsible for several epidemics worldwide; however, the effective antiviral drug is unavailable to date. The 2A proteinase (2A(pro)) of EV71 presents a promising drug target due to its multiple roles in virus replication, inhibition of host protein synthesis and evasion of innate immunity. We determined the crystal structure of EV71 2A(pro) at 1.85Å resolution, revealing that the proteinase maintains a chymotrypsin-like fold. The active site is composed of the catalytic triads C110A, H21 and D39 with the geometry similar to that in other picornaviral 2A(pro), 3C(pro) and serine proteinases. The cI-to-eI2 loop at the N-terminal domain of EV71 2A(pro) adopts a highly stable conformation and contributes to the hydrophilic surface property, which are strikingly different in HRV2 2A(pro) but are similar in CVB4 2A(pro). We identified a hydrophobic motif "LLWL" followed by an acidic motif "DEE" at the C-terminus of EV71 2A(pro). The "LLWL" motif is folded into the ß-turn structure that is essential for the positioning of the acidic motif. Our structural and mutagenesis study demonstrated that both the negative charging and the correct positioning of the C-terminus are essential for EV71 replication. Deletion of the "LLWL" motif abrogated the proteolytic activity, indicating that the motif is critical for maintaining the active proteinase conformation. Our findings provide the structural and functional insights into EV71 2A(pro) and establish a framework for structure-based inhibitor design.


Assuntos
Cisteína Endopeptidases/química , Vírus da Febre Aftosa/enzimologia , Proteínas Virais/química , Sequência de Aminoácidos , Domínio Catalítico , Vírus da Febre Aftosa/genética , Ligação de Hidrogênio , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Alinhamento de Sequência , Replicação Viral
14.
Virology ; 443(2): 271-7, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23756127

RESUMO

The foot-and-mouth disease virus leader proteinase (Lb(pro)) cleaves itself off the nascent viral polyprotein. NMR studies on the monomeric variant Lb(pro) L200F provide structural evidence for intramolecular self-processing. (15)N-HSQC measurements of Lb(pro) L200F showed specifically shifted backbone signals in the active and substrate binding sites compared to the monomeric variant sLb(pro), lacking six C-terminal residues. This indicates transient intramolecular interactions between the C-terminal extension (CTE) of one molecule and its own active site. Contrastingly, the porcine reproductive and respiratory syndrome virus (PRRSV) leader proteinase nsp1α, with a papain-like fold like Lb(pro), stably binds its own CTE. Parts of the ß-sheet domains but none of the α-helical domains of Lb(pro) and nsp1α superimpose; consequently, the α-helical domain of nsp1α is oriented differently relative to its ß-sheet domain. This provides a large interaction surface for the CTE with the globular domain, stabilising the intramolecular complex. Consequently, self-processing inactivates nsp1α but not Lb(pro).


Assuntos
Endopeptidases/metabolismo , Vírus da Febre Aftosa/enzimologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/enzimologia , Proteínas não Estruturais Virais/metabolismo , Animais , Sítios de Ligação , Domínio Catalítico , Endopeptidases/química , Endopeptidases/genética , Vírus da Febre Aftosa/genética , Modelos Moleculares , Mutação , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Dobramento de Proteína , Relação Estrutura-Atividade , Especificidade por Substrato , Suínos/virologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética
15.
Infect Genet Evol ; 16: 270-4, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23499709

RESUMO

The 3C protease of foot-and-mouth disease virus (FMDV) has a conserved amino acid sequence and is responsible for most cleavage in the viral polyprotein. The effects of the synonymous codon usage of FMDV 3C gene and tRNA abundance of the hosts on shaping different folding units (α-helix, ß-strand and the coil) in the 3C protease were analyzed based on the structural information of the FMDV 3C protease from Protein Data Bank (PDB: 2BHG) and 210 genes of 3C for all serotypes of FMDV. The strong correlation between some codons usage and the specific folding unit in the FMDV 3C protease is found. As for the effect of translation speed caused by tRNA abundance on the formation of folding units, the codon positions with lowly abundant tRNA scatter in the 3C gene and there is the obvious fluctuation of tRNA abundance locating in the transition boundaries from the ß-strand to the α-helix and the coil, respectively. However, codon positions with lowly abundant tRNA clustering into these boundaries are not found, suggesting that the adjustment of translation speed is likely also achieved by the single codon position with low tRNA abundance rather than a cluster. The observations can provide the information for insight into the role of the synonymous codon usage in the formation of 3C protease of FMDV and effect of the tRNA abundance of the hosts on this formation of protease.


Assuntos
Cisteína Endopeptidases/genética , Vírus da Febre Aftosa/genética , Dobramento de Proteína , RNA de Transferência/genética , Proteínas Virais/genética , Proteases Virais 3C , Sequência de Aminoácidos , Aminoácidos/genética , Sequência de Bases , Códon , Sequência Conservada/genética , Cisteína Endopeptidases/metabolismo , Vírus da Febre Aftosa/enzimologia , Proteínas Virais/metabolismo
16.
J Gen Virol ; 94(Pt 6): 1249-1258, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23364188

RESUMO

The foot-and-mouth disease virus (FMDV) capsid protein precursor (P1-2A) is processed by the virus-encoded 3C protease (3C(pro)) to produce VP0, VP3, VP1 and 2A. Within the virus-encoded polyprotein, the P1-2A and 3C(pro) can be expected to be produced at equivalent concentrations. However, using transient-expression assays, within mammalian cells, it is possible to modify the relative amounts of the substrate and protease. It has now been shown that optimal production of the processed capsid proteins from P1-2A is achieved with reduced levels of 3C(pro) expression, relative to the P1-2A, compared with that achieved with a single P1-2A-3C polyprotein. Expression of the FMDV 3C(pro) is poorly tolerated by mammalian cells and higher levels of the 3C(pro) greatly inhibit protein expression. In addition, it is demonstrated that both the intact P1-2A precursor and the processed capsid proteins can be efficiently detected by FMDV antigen detection assays. Furthermore, the P1-2A and the processed forms each bind to the integrin αvß6, the major FMDV receptor. These results contribute to the development of systems which efficiently express the components of empty capsid particles and may represent the basis for safer production of diagnostic reagents and improved vaccines against foot-and-mouth disease.


Assuntos
Proteínas do Capsídeo/genética , Cisteína Endopeptidases/genética , Vírus da Febre Aftosa/enzimologia , Febre Aftosa/virologia , Regulação Viral da Expressão Gênica , Proteínas Virais/genética , Proteases Virais 3C , Animais , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Cricetinae , Cisteína Endopeptidases/metabolismo , Vírus da Febre Aftosa/genética , Processamento de Proteína Pós-Traducional , Proteínas Virais/metabolismo
17.
J Virol ; 86(17): 9311-22, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22718831

RESUMO

Foot-and-mouth disease is a highly contagious viral illness of wild and domestic cloven-hoofed animals. The causative agent, foot-and-mouth disease virus (FMDV), replicates rapidly, efficiently disseminating within the infected host and being passed on to susceptible animals via direct contact or the aerosol route. To survive in the host, FMDV has evolved to block the host interferon (IFN) response. Previously, we and others demonstrated that the leader proteinase (L(pro)) of FMDV is an IFN antagonist. Here, we report that another FMDV-encoded proteinase, 3C(pro), also inhibits IFN-α/ß response and the expression of IFN-stimulated genes. Acting in a proteasome- and caspase-independent manner, the 3C(pro) of FMDV proteolytically cleaved nuclear transcription factor kappa B (NF-κB) essential modulator (NEMO), a bridging adaptor protein essential for activating both NF-κB and interferon-regulatory factor signaling pathways. 3C(pro) specifically targeted NEMO at the Gln 383 residue, cleaving off the C-terminal zinc finger domain from the protein. This cleavage impaired the ability of NEMO to activate downstream IFN production and to act as a signaling adaptor of the RIG-I/MDA5 pathway. Mutations specifically disrupting the cysteine protease activity of 3C(pro) abrogated NEMO cleavage and the inhibition of IFN induction. Collectively, our data identify NEMO as a substrate for FMDV 3C(pro) and reveal a novel mechanism evolved by a picornavirus to counteract innate immune signaling.


Assuntos
Cisteína Endopeptidases/metabolismo , Vírus da Febre Aftosa/enzimologia , Febre Aftosa/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Transdução de Sinais , Doenças dos Suínos/imunologia , Proteínas Virais/metabolismo , Proteases Virais 3C , Animais , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/imunologia , Febre Aftosa/genética , Febre Aftosa/metabolismo , Febre Aftosa/virologia , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/imunologia , Imunidade Inata , Interferons/genética , Interferons/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , NF-kappa B/genética , NF-kappa B/imunologia , Proteólise , Suínos , Doenças dos Suínos/genética , Doenças dos Suínos/metabolismo , Doenças dos Suínos/virologia , Proteínas Virais/genética , Proteínas Virais/imunologia
18.
Virology ; 425(1): 40-52, 2012 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-22280896

RESUMO

Picornavirus infection can lead to disruption of nuclear pore traffic, shut-off of cell translation machinery, and cleavage of proteins involved in cellular signal transduction and the innate response to infection. Here, we demonstrated that the FMDV 3C(pro) induced the cleavage of nuclear RNA-binding protein Sam68 C-terminus containing the nuclear localization sequence (NLS). Consequently, it stimulated the redistribution of Sam68 to the cytoplasm. The siRNA knockdown of Sam68 resulted in a 1000-fold reduction in viral titers, which prompted us to study the effect of Sam68 on FMDV post-entry events. Interestingly, Sam68 interacts with the internal ribosomal entry site within the 5' non-translated region of the FMDV genome, and Sam68 knockdown decreased FMDV IRES-driven activity in vitro suggesting that it could modulate translation of the viral genome. The results uncover a novel role for Sam68 in the context of picornaviruses and the proteolysis of a new cellular target of the FMDV 3C(pro).


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cisteína Endopeptidases/metabolismo , Citoplasma/metabolismo , Vírus da Febre Aftosa/patogenicidade , Rim/virologia , Proteínas de Ligação a RNA/metabolismo , Proteínas Virais/metabolismo , Proteases Virais 3C , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Cricetinae , Vírus da Febre Aftosa/enzimologia , Rim/citologia , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Biossíntese de Proteínas , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Ribossomos/virologia
19.
J Gen Virol ; 93(Pt 3): 504-515, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22131310

RESUMO

Mature protein C of tick-borne encephalitis virus (TBEV) is cleaved from the polyprotein precursor by the viral NS2B/3 protease (NS2B/3(pro)). We showed previously that replacement of the NS2B/3(pro) cleavage site at the C terminus of protein C by the foot-and-mouth disease virus (FMDV) 2A StopGo sequence leads to the production of infectious virions. Here, we show that infectious virions can also be produced from a TBEV mutant bearing an inactivated 2A sequence through the expression of the FMDV 3C protease (3C(pro)) either in cis or in trans (from a TBEV replicon). Cleavage at the C terminus of protein C depended on the catalytic activity of 3C(pro) as well as on the presence of an optimized 3C(pro) cleavage site. Passage of the TBEV mutants bearing a 3C(pro) cleavage site either in the absence of 3C(pro) or in the presence of a catalytically inactive 3C(pro) led to the appearance of revertants in which protein C cleavage by NS2B/3(pro) had been regained. In three different revertants, a cleavage site for NS2B/3(pro), namely RR*C, was now present, leading to an elongated protein C. Furthermore, two revertants acquired additional mutations in the C terminus of protein C, eliminating two basic residues. Although these latter mutants showed wild-type levels of early RNA synthesis, their foci were smaller and an accumulation of protein C in the cytoplasm was observed. These findings suggest a role of the positive charge of the C terminus of protein C for budding of the nucleocapsid and further support the notion that TBEV protein C is a multifunctional protein.


Assuntos
Cisteína Endopeptidases/metabolismo , Vírus da Encefalite Transmitidos por Carrapatos/fisiologia , Vírus da Febre Aftosa/enzimologia , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Proteases Virais 3C , Cisteína Endopeptidases/genética , Vírus da Encefalite Transmitidos por Carrapatos/genética , Mutação , RNA Helicases/genética , RNA Helicases/metabolismo , Recombinação Genética , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética
20.
J Mol Biol ; 395(2): 375-89, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19883658

RESUMO

Picornavirus replication is critically dependent on the correct processing of a polyprotein precursor by 3C protease(s) (3C(pro)) at multiple specific sites with related but non-identical sequences. To investigate the structural basis of its cleavage specificity, we performed the first crystallographic structural analysis of non-covalent complexes of a picornavirus 3C(pro) with peptide substrates. The X-ray crystal structure of the foot-and-mouth disease virus 3C(pro), mutated to replace the catalytic Cys by Ala and bound to a peptide (APAKQ|LLNFD) corresponding to the P5-P5' region of the VP1-2A cleavage junction in the viral polyprotein, was determined up to 2.5 A resolution. Comparison with free enzyme reveals significant conformational changes in 3C(pro) on substrate binding that lead to the formation of an extended interface of contact primarily involving the P4-P2' positions of the peptide. Strikingly, the deep S1' specificity pocket needed to accommodate P1'-Leu only forms when the peptide binds. Substrate specificity was investigated using peptide cleavage assays to show the impact of amino acid substitutions within the P5-P4' region of synthetic substrates. The structure of the enzyme-peptide complex explains the marked substrate preferences for particular P4, P2 and P1 residue types, as well as the relative promiscuity at P3 and on the P' side of the scissile bond. Furthermore, crystallographic analysis of the complex with a modified VP1-2A peptide (APAKE|LLNFD) containing a Gln-to-Glu substitution reveals an identical mode of peptide binding and explains the ability of foot-and-mouth disease virus 3C(pro) to cleave sequences containing either P1-Gln or P1-Glu. Structure-based mutagenesis was used to probe interactions within the S1' specificity pocket and to provide direct evidence of the important contribution made by Asp84 of the Cys-His-Asp catalytic triad to proteolytic activity. Our results provide a new level of detail in our understanding of the structural basis of polyprotein cleavage by 3C(pro).


Assuntos
Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Vírus da Febre Aftosa/enzimologia , Proteínas Virais/química , Proteínas Virais/metabolismo , Proteases Virais 3C , Sequência de Aminoácidos , Substituição de Aminoácidos , Sequência de Bases , Domínio Catalítico/genética , Cristalografia por Raios X , Cisteína Endopeptidases/genética , DNA Viral/genética , Vírus da Febre Aftosa/genética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Peptídeos/química , Peptídeos/genética , Ligação Proteica , Conformação Proteica , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Proteínas Virais/genética , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA