Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
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.
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
3.
J Virol ; 93(2)2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30404792

RESUMO

Like other viruses, the picornavirus foot-and-mouth disease virus (FMDV; genus Aphthovirus), one of the most notorious pathogens in the global livestock industry, needs to navigate antiviral host responses to establish an infection. There is substantial insight into how FMDV suppresses the type I interferon (IFN) response, but it is largely unknown whether and how FMDV modulates the integrated stress response. Here, we show that the stress response is suppressed during FMDV infection. Using a chimeric recombinant encephalomyocarditis virus (EMCV), in which we functionally replaced the endogenous stress response antagonist by FMDV leader protease (Lpro) or 3Cpro, we demonstrate an essential role for Lpro in suppressing stress granule (SG) formation. Consistently, infection with a recombinant FMDV lacking Lpro resulted in SG formation. Additionally, we show that Lpro cleaves the known SG scaffold proteins G3BP1 and G3BP2 but not TIA-1. We demonstrate that the closely related equine rhinitis A virus (ERAV) Lpro also cleaves G3BP1 and G3BP2 and also suppresses SG formation, indicating that these abilities are conserved among aphthoviruses. Neither FMDV nor ERAV Lpro interfered with phosphorylation of RNA-dependent protein kinase (PKR) or eIF2α, indicating that Lpro does not affect SG formation by inhibiting the PKR-triggered signaling cascade. Taken together, our data suggest that aphthoviruses actively target scaffolding proteins G3BP1 and G3BP2 and antagonize SG formation to modulate the integrated stress response.IMPORTANCE The picornavirus foot-and-mouth disease virus (FMDV) is a notorious animal pathogen that puts a major economic burden on the global livestock industry. Outbreaks have significant consequences for animal health and product safety. Like many other viruses, FMDV must manipulate antiviral host responses to establish infection. Upon infection, viral double-stranded RNA (dsRNA) is detected, which results in the activation of the RNA-dependent protein kinase (PKR)-mediated stress response, leading to a stop in cellular and viral translation and the formation of stress granules (SG), which are thought to have antiviral properties. Here, we show that FMDV can suppress SG formation via its leader protease (Lpro). Simultaneously, we observed that Lpro can cleave the SG scaffolding proteins G3BP1 and G3BP2. Understanding the molecular mechanisms of the antiviral host response evasion strategies of FMDV may help to develop countermeasures to control FMDV infections in the future.


Assuntos
Grânulos Citoplasmáticos/metabolismo , Vírus da Febre Aftosa/enzimologia , Febre Aftosa/metabolismo , Peptídeo Hidrolases/metabolismo , Proteínas com Motivo de Reconhecimento de RNA/metabolismo , Animais , Aphthovirus/enzimologia , Linhagem Celular , Cricetinae , Vírus da Encefalomiocardite/enzimologia , Febre Aftosa/virologia , Células HEK293 , Células HeLa , Humanos , Estresse Fisiológico , Proteínas Virais/metabolismo
4.
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
5.
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
6.
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
7.
J Virol ; 92(20)2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30068642

RESUMO

Viral RNA-dependent RNA polymerases (RdRps) are major determinants of high mutation rates and generation of mutant spectra that mediate RNA virus adaptability. The RdRp of the picornavirus foot-and-mouth disease virus (FMDV), termed 3D, is a multifunctional protein that includes a nuclear localization signal (NLS) in its N-terminal region. Previous studies documented that some amino acid substitutions within the NLS altered nucleotide recognition and enhanced the incorporation of the mutagenic purine analogue ribavirin in viral RNA, but the mutants tested were not viable and their response to lethal mutagenesis could not be studied. Here we demonstrate that NLS amino acid substitution M16A of FMDV serotype C does not affect infectious virus production but accelerates ribavirin-mediated virus extinction. The mutant 3D displays polymerase activity, RNA binding, and copying processivity that are similar to those of the wild-type enzyme but shows increased ribavirin-triphosphate incorporation. Crystal structures of the mutant 3D in the apo and RNA-bound forms reveal an expansion of the template entry channel due to the replacement of the bulky Met by Ala. This is a major difference with other 3D mutants with altered nucleotide analogue recognition. Remarkably, two distinct loop ß9-α11 conformations distinguish 3Ds that exhibit higher or lower ribavirin incorporation than the wild-type enzyme. This difference identifies a specific molecular determinant of ribavirin sensitivity of FMDV. Comparison of several polymerase mutants indicates that different domains of the molecule can modify nucleotide recognition and response to lethal mutagenesis. The connection of this observation with current views on quasispecies adaptability is discussed.IMPORTANCE The nuclear localization signal (NLS) of the foot-and-mouth disease virus (FMDV) polymerase includes residues that modulate the sensitivity to mutagenic agents. Here we have described a viable NLS mutant with an amino acid replacement that facilitates virus extinction by ribavirin. The corresponding polymerase shows increased incorporation of ribavirin triphosphate and local structural modifications that implicate the template entry channel. Specifically, comparison of the structures of ribavirin-sensitive and ribavirin-resistant FMDV polymerases has identified loop ß9-α11 conformation as a determinant of sensitivity to ribavirin mutagenesis.


Assuntos
Vírus da Febre Aftosa/enzimologia , Mutagênese , RNA Polimerase Dependente de RNA/metabolismo , Substituição de Aminoácidos , Animais , Antivirais/metabolismo , Linhagem Celular , Cricetinae , Cristalografia por Raios X , Sinais de Localização Nuclear , RNA Viral/biossíntese , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/genética , Ribavirina/metabolismo
8.
J Virol ; 91(15)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28515297

RESUMO

Foot-and-mouth disease virus (FMDV) RNA-dependent RNA polymerase (RdRp) (3Dpol) catalyzes viral RNA synthesis. Its characteristic low fidelity and absence of proofreading activity allow FMDV to rapidly mutate and adapt to dynamic environments. In this study, we used the structure of FMDV 3Dpol in combination with previously reported results from similar picornaviral polymerases to design point mutations that would alter replication fidelity. In particular, we targeted Trp237 within conserved polymerase motif A because of the low reversion potential inherent in the single UGG codon. Using biochemical and genetic tools, we show that the replacement of tryptophan 237 with phenylalanine imparts higher fidelity, but replacements with isoleucine and leucine resulted in lower-fidelity phenotypes. Viruses containing these W237 substitutions show in vitro growth kinetics and plaque morphologies similar to those of the wild-type (WT) A24 Cruzeiro strain in BHK cells, and both high- and low-fidelity variants retained fitness during coinfection with the wild-type virus. The higher-fidelity W237F (W237FHF) mutant virus was more resistant to the mutagenic nucleoside analogs ribavirin and 5-fluorouracil than the WT virus, whereas the lower-fidelity W237I (W237ILF) and W237LLF mutant viruses exhibited lower ribavirin resistance. Interestingly, the variant viruses showed heterogeneous and slightly delayed growth kinetics in primary porcine kidney cells, and they were significantly attenuated in mouse infection experiments. These data demonstrate, for a single virus, that either increased or decreased RdRp fidelity attenuates virus growth in animals, which is a desirable feature for the development of safer and genetically more stable vaccine candidates.IMPORTANCE Foot-and-mouth disease (FMD) is the most devastating disease affecting livestock worldwide. Here, using structural and biochemical analyses, we have identified FMDV 3Dpol mutations that affect polymerase fidelity. Recombinant FMDVs containing substitutions at 3Dpol tryptophan residue 237 were genetically stable and displayed plaque phenotypes and growth kinetics similar to those of the wild-type virus in cell culture. We further demonstrate that viruses harboring either a W237FHF substitution or W237ILF and W237LLF mutations were highly attenuated in animals. Our study shows that obtaining 3Dpol fidelity variants by protein engineering based on polymerase structure and function could be exploited for the development of attenuated FMDV vaccine candidates that are safer and more stable than strains obtained by selective pressure via mutagenic nucleotides or adaptation approaches.


Assuntos
Antígenos Virais/genética , Antígenos Virais/metabolismo , Vírus da Febre Aftosa/enzimologia , Vírus da Febre Aftosa/patogenicidade , Engenharia de Proteínas , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Substituição de Aminoácidos , Animais , Antivirais , Células Cultivadas , Cricetinae , Análise Mutacional de DNA , Modelos Animais de Doenças , Farmacorresistência Viral , Fluoruracila/farmacologia , Febre Aftosa/patologia , Febre Aftosa/virologia , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/crescimento & desenvolvimento , Camundongos , Mutagênese Sítio-Dirigida , Mutação Puntual , Ribavirina/farmacologia , Suínos , Triptofano/genética , Triptofano/metabolismo , Ensaio de Placa Viral
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 Virol ; 90(15): 6864-6883, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27194768

RESUMO

UNLABELLED: The Picornaviridae is a large family of positive-sense RNA viruses that contains numerous human and animal pathogens, including foot-and-mouth disease virus (FMDV). The picornavirus replication complex comprises a coordinated network of protein-protein and protein-RNA interactions involving multiple viral and host-cellular factors. Many of the proteins within the complex possess multiple roles in viral RNA replication, some of which can be provided in trans (i.e., via expression from a separate RNA molecule), while others are required in cis (i.e., expressed from the template RNA molecule). In vitro studies have suggested that multiple copies of the RNA-dependent RNA polymerase (RdRp) 3D are involved in the viral replication complex. However, it is not clear whether all these molecules are catalytically active or what other function(s) they provide. In this study, we aimed to distinguish between catalytically active 3D molecules and those that build a replication complex. We report a novel nonenzymatic cis-acting function of 3D that is essential for viral-genome replication. Using an FMDV replicon in complementation experiments, our data demonstrate that this cis-acting role of 3D is distinct from the catalytic activity, which is predominantly trans acting. Immunofluorescence studies suggest that both cis- and trans-acting 3D molecules localize to the same cellular compartment. However, our genetic and structural data suggest that 3D interacts in cis with RNA stem-loops that are essential for viral RNA replication. This study identifies a previously undescribed aspect of picornavirus replication complex structure-function and an important methodology for probing such interactions further. IMPORTANCE: Foot-and-mouth disease virus (FMDV) is an important animal pathogen responsible for foot-and-mouth disease. The disease is endemic in many parts of the world with outbreaks within livestock resulting in major economic losses. Propagation of the viral genome occurs within replication complexes, and understanding this process can facilitate the development of novel therapeutic strategies. Many of the nonstructural proteins involved in replication possess multiple functions in the viral life cycle, some of which can be supplied to the replication complex from a separate genome (i.e., in trans) while others must originate from the template (i.e., in cis). Here, we present an analysis of cis and trans activities of the RNA-dependent RNA polymerase 3D. We demonstrate a novel cis-acting role of 3D in replication. Our data suggest that this role is distinct from its enzymatic functions and requires interaction with the viral genome. Our data further the understanding of genome replication of this important pathogen.


Assuntos
Antígenos Virais/metabolismo , Vírus da Febre Aftosa/enzimologia , Vírus da Febre Aftosa/genética , Febre Aftosa/virologia , RNA Viral/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/genética , Animais , Antígenos Virais/química , Células Cultivadas , Genoma Viral , Humanos , Modelos Moleculares , Conformação Proteica , Transcrição Gênica , Proteínas não Estruturais Virais/química
11.
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
12.
J Virol ; 89(13): 6848-59, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25903341

RESUMO

UNLABELLED: The N-terminal region of the foot-and-mouth disease virus (FMDV) 3D polymerase contains the sequence MRKTKLAPT (residues 16 to 24) that acts as a nuclear localization signal. A previous study showed that substitutions K18E and K20E diminished the transport to the nucleus of 3D and 3CD and severely impaired virus infectivity. These residues have also been implicated in template binding, as seen in the crystal structures of different 3D-RNA elongation complexes. Here, we report the biochemical and structural characterization of different mutant polymerases harboring substitutions at residues 18 and 20, in particular, K18E, K18A, K20E, K20A, and the double mutant K18A K20A (KAKA). All mutant enzymes exhibit low RNA binding activity, low processivity, and alterations in nucleotide recognition, including increased incorporation of ribavirin monophosphate (RMP) relative to the incorporation of cognate nucleotides compared with the wild-type enzyme. The structural analysis shows an unprecedented flexibility of the 3D mutant polymerases, including both global rearrangements of the closed-hand architecture and local conformational changes at loop ß9-α11 (within the polymerase motif B) and at the template-binding channel. Specifically, in 3D bound to RNA, both K18E and K20E induced the opening of new pockets in the template channel where the downstream templating nucleotide at position +2 binds. The comparisons of free and RNA-bound enzymes suggest that the structural rearrangements may occur in a concerted mode to regulate RNA replication, processivity, and fidelity. Thus, the N-terminal region of FMDV 3D that acts as a nuclear localization signal (NLS) and in template binding is also involved in nucleotide recognition and can affect the incorporation of nucleotide analogues. IMPORTANCE: The study documents multifunctionality of a nuclear localization signal (NLS) located at the N-terminal region of the foot-and-mouth disease viral polymerase (3D). Amino acid substitutions at this polymerase region can impair the transport of 3D to the nucleus, reduce 3D binding to RNA, and alter the relative incorporation of standard nucleoside monophosphate versus ribavirin monophosphate. Structural data reveal that the conformational changes in this region, forming part of the template channel entry, would be involved in nucleotide discrimination. The results have implications for the understanding of viral polymerase function and for lethal mutagenesis mechanisms.


Assuntos
Antígenos Virais/química , Antígenos Virais/metabolismo , Vírus da Febre Aftosa/enzimologia , Sinais de Localização Nuclear , Nucleotídeos/metabolismo , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Substituição de Aminoácidos , Antígenos Virais/genética , Cristalografia por Raios X , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Ligação Proteica , Conformação Proteica , RNA/metabolismo , Proteínas não Estruturais Virais/genética
13.
J Virol ; 88(8): 4008-20, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24453363

RESUMO

UNLABELLED: Mutagenic nucleoside analogues can be used to isolate RNA virus high-fidelity RNA-dependent RNA polymerase (RdRp) variants, the majority of which are attenuated in vivo. However, attenuated foot-and-mouth disease virus (FMDV) high-fidelity RdRp variants have not been isolated, and the correlations between RdRp fidelity and virulence remain unclear. Here, the mutagen ribavirin was used to select a ribavirin-resistant population of FMDV, and 4 amino acid substitutions (D5N, A38V, M194I, and M296V) were identified in the RdRp-coding region of the population. Through single or combined mutagenesis using a reverse genetics system, we generated direct experimental evidence that the rescued D5N, A38V, and DAMM mutants but not the M194I and M296V mutants are high-fidelity RdRp variants. Mutagen resistance assays revealed that the higher replication fidelity was associated with higher-level resistance to ribavirin. In addition, significantly attenuated fitness and virulence phenotypes were observed for the D5N, A38V, and DAMM mutants. Based on a systematic quantitative analysis of fidelity and virulence, we concluded that higher replication fidelity is associated with a more attenuated virus. These data suggest that the resulting restricted quasispecies diversity compromises the adaptability and virulence of an RNA virus population. The modulation of replication fidelity to attenuate virulence may represent a general strategy for the rational design of new types of live, attenuated vaccine strains. IMPORTANCE: The ribavirin-isolated poliovirus (PV) RdRp G64S variant, the polymerases of which were of high replication fidelity, was attenuated in vivo. It has been proposed (M. Vignuzzi, E. Wendt, and R. Andino, Nat. Med. 14:154-161, http://dx.doi.org/10.1038/nm1726) that modulation of replication fidelity is a promising approach for engineering attenuated virus vaccines. The subsequently mutagen-isolated RdRp variants also expressed the high-fidelity polymerase, but not all of them were attenuated. Few studies have shown the exact correlation between fidelity and virulence. The present study investigates the effect of restricted quasispecies diversity on viral virulence via several attenuated FMDV high-fidelity RdRp variants. Our findings may aid in the rational design of a new type of vaccine strain.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral , Vírus da Febre Aftosa/efeitos dos fármacos , Vírus da Febre Aftosa/patogenicidade , Febre Aftosa/virologia , Variação Genética , Ribavirina/farmacologia , Animais , Sequência de Bases , Feminino , Vírus da Febre Aftosa/enzimologia , Vírus da Febre Aftosa/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Mutação de Sentido Incorreto , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Virulência/efeitos dos fármacos
14.
Biol Chem ; 395(10): 1179-85, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24670358

RESUMO

The leader proteinase (Lpro) of the foot-and-mouth disease virus inhibits the host innate immune response by at least three different mechanisms. The most well-characterised of these is the prevention of the synthesis of cytokines such as interferons immediately after infection, brought about by specific proteolytic cleavage of the eukaryotic initiation factor 4G. This prevents the recruitment of capped cellular mRNA; however, the viral RNA can be translated under these conditions. The two other mechanisms are the induction of NF-κB cleavage and the deubiquitination of immune signalling molecules. This review focuses on the structure-function relationships in Lpro responsible for these widely divergent activities.


Assuntos
Vírus da Febre Aftosa/enzimologia , Vírus da Febre Aftosa/genética , Proteínas de Membrana/metabolismo , Serina Endopeptidases/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Animais , Proteínas de Membrana/genética , Modelos Moleculares , Estrutura Secundária de Proteína , Inibidores da Síntese de Proteínas , Serina Endopeptidases/genética
15.
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
16.
Arch Virol ; 159(10): 2641-50, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24888311

RESUMO

Previous studies have shown that RNA viruses can be attenuated by either increased or decreased viral polymerase replication fidelity. Although foot-and-mouth disease virus (FMDV) high-fidelity RNA-dependent RNA polymerase (RdRp) variants with an attenuated phenotype have been isolated using mutagens, no FMDV mutant with a low-fidelity polymerase has been documented to date. Here, we describe the generation of several FMDV RdRp mutants using site-directed mutagenesis via a reverse genetic system. Mutation frequency assays confirmed that five rescued FMDV RdRp mutant populations had lower replication fidelity than the wild-type virus population, which allowed us to assess the effects of the change in replication fidelity on the virus phenotype. These low-fidelity FMDV RdRp mutants showed increased sensitivity to ribavirin or 5-fluorouracil (5-FU) treatment without a loss of growth capacity in cell cultures. In addition, decreased fitness and attenuated virulence were observed for the RdRp mutants with lower fidelity. Importantly, based on a quantitative analysis for fidelity and virulence, we concluded that lower replication fidelity is associated with a more attenuated virus phenotype. These results further contribute to our understanding of the replication fidelity of polymerases of RNA viruses and its relationship to virulence attenuation.


Assuntos
Vírus da Febre Aftosa/enzimologia , Vírus da Febre Aftosa/genética , RNA Polimerase Dependente de RNA/genética , Replicação Viral/genética , Animais , Antivirais/farmacologia , Sequência de Bases , Linhagem Celular , Cricetinae , Farmacorresistência Viral/genética , Fluoruracila/farmacologia , Febre Aftosa/virologia , Vírus da Febre Aftosa/efeitos dos fármacos , Vírus da Febre Aftosa/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Taxa de Mutação , RNA Viral/biossíntese , RNA Viral/genética , Ribavirina/farmacologia , Análise de Sequência de RNA
17.
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
18.
J Virol ; 86(4): 2371-4, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22156531

RESUMO

The replication of many viruses involves the formation of higher-order structures or replication "factories." We show that the key replication enzyme of foot-and-mouth disease virus (FMDV), the RNA-dependent RNA polymerase, forms fibrils in vitro. Although there are similarities with previously characterized poliovirus polymerase fibrils, FMDV fibrils are narrower, are composed of both protein and RNA, and, importantly, are seen only when all components of an elongation assay are present. Furthermore, an inhibitory RNA aptamer prevents fibril formation.


Assuntos
Replicação do DNA , Vírus da Febre Aftosa/enzimologia , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/química , Linhagem Celular , Vírus da Febre Aftosa/química , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/fisiologia , RNA Viral/genética , RNA Viral/metabolismo , RNA Polimerase Dependente de RNA/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA