Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 295(6): 1694-1703, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-31914414

RESUMO

Small-molecule inhibitors of translation are critical tools to study the molecular mechanisms of protein synthesis. In this study, we sought to characterize how QL47, a host-targeted, small-molecule antiviral agent, inhibits steady-state viral protein expression. We demonstrate that this small molecule broadly inhibits both viral and host protein synthesis and targets a translation step specific to eukaryotic cells. We show that QL47 inhibits protein neosynthesis initiated by both canonical cap-driven and noncanonical initiation strategies, most likely by targeting an early step in translation elongation. Our findings thus establish QL47 as a new small-molecule inhibitor that can be utilized to probe the eukaryotic translation machinery and that can be further developed as a new therapeutic agent.


Assuntos
Antivirais/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Antivirais/química , Linhagem Celular , Células HEK293 , Humanos , Proteínas/metabolismo , Bibliotecas de Moléculas Pequenas/química , Proteínas Virais/metabolismo , Viroses/tratamento farmacológico , Viroses/metabolismo , Vírus/efeitos dos fármacos , Vírus/metabolismo
2.
Phytopathology ; 110(1): 228-236, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31411546

RESUMO

A hallmark feature of (+)-strand RNA viruses of eukaryotic cells is that progeny (+)-strands are accumulated 100-fold over (-)-strands. Previous experimental evidence suggests that, in Brome mosaic virus (BMV), a plant-infecting member of the alphavirus-like superfamily, the addition of RNA3 and, specifically, translation of the wild-type (WT) coat protein (CP) gene contributes to increased accumulation of (+)-strands. It is unclear whether this stimulation of (+)-strand accumulation by CP is due to direct regulation of viral RNA replication or RNA stabilization via encapsidation. Analysis of BMV progeny RNA in Nicotiana benthamiana plants revealed that expression of RNA3 variants that did not express WT CP led to a severe defect in BMV (+)-strand accumulation. The (+)-strand accumulation could be rescued when CP was complemented in trans. To verify whether stimulation of (+)-strand accumulation is coupled with encapsidation, two independent mutations were engineered into CP open reading frames. An N-terminal deletion that prevented CP binding to the viral RNAs resulted in a severe reduction of BMV (+)-strand accumulation but stimulated (-)-strand accumulation over the WT. On the other hand, a C-terminal mutation affecting CP dimerization caused a significant decrease in (+)-strand accumulation but had no detectable effect on (-)-strand accumulation. Nucleotide sequences in the movement protein-coding region were also found to contribute to (+)-strand accumulation, in part by providing packaging signals for efficient RNA3 encapsidation. Overall, these results show that RNA encapsidation is a significant determinant of BMV RNA intracellular accumulation.


Assuntos
Bromovirus , Proteínas do Capsídeo , Nicotiana , Doenças das Plantas , Bromovirus/fisiologia , Proteínas do Capsídeo/genética , Doenças das Plantas/virologia , RNA Viral/genética , Nicotiana/virologia , Replicação Viral
3.
Nat Commun ; 10(1): 3468, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31371704

RESUMO

Targeted protein degradation is a promising drug development paradigm. Here we leverage this strategy to develop a new class of small molecule antivirals that induce proteasomal degradation of viral proteins. Telaprevir, a reversible-covalent inhibitor that binds to the hepatitis C virus (HCV) protease active site is conjugated to ligands that recruit the CRL4CRBN ligase complex, yielding compounds that can both inhibit and induce the degradation of the HCV NS3/4A protease. An optimized degrader, DGY-08-097, potently inhibits HCV in a cellular infection model, and we demonstrate that protein degradation contributes to its antiviral activity. Finally, we show that this new class of antiviral agents can overcome viral variants that confer resistance to traditional enzymatic inhibitors such as telaprevir. Overall, our work provides proof-of-concept that targeted protein degradation may provide a new paradigm for the development of antivirals with superior resistance profiles.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antivirais/química , Linhagem Celular Tumoral , Desenho de Fármacos , Farmacorresistência Viral/genética , Técnicas de Silenciamento de Genes , Células HEK293 , Hepacivirus/efeitos dos fármacos , Hepacivirus/metabolismo , Hepatite C/tratamento farmacológico , Hepatite C/genética , Hepatite C/virologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ligantes , Modelos Moleculares , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Estudo de Prova de Conceito , Inibidores de Proteases/química , Proteólise/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo , Proteínas não Estruturais Virais/metabolismo
4.
ACS Infect Dis ; 5(3): 460-472, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30608640

RESUMO

Vaccines and antivirals to combat dengue, Zika, and other flavivirus pathogens present a major, unmet medical need. Vaccine development has been severely challenged by the antigenic diversity of these viruses and the propensity of non-neutralizing, cross-reactive antibodies to facilitate cellular infection and increase disease severity. As an alternative, direct-acting antivirals targeting the flavivirus envelope protein, E, have the potential to act via an analogous mode of action without the risk of antibody-dependent enhancement of infection and disease. We previously discovered that structurally diverse small molecule inhibitors of the dengue virus E protein exhibit varying levels of antiviral activity against other flaviviruses in cell culture. Here, we demonstrate that the broad-spectrum activity of several cyanohydrazones against dengue, Zika, and Japanese encephalitis viruses is due to specific inhibition of E-mediated membrane fusion during viral entry and provide proof of concept for pharmacological inhibition of E as an antiviral strategy in vivo.


Assuntos
Antivirais/administração & dosagem , Infecções por Flavivirus/tratamento farmacológico , Flavivirus/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/administração & dosagem , Proteínas do Envelope Viral/metabolismo , Animais , Antivirais/química , Feminino , Flavivirus/fisiologia , Infecções por Flavivirus/virologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Bibliotecas de Moléculas Pequenas/química , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/genética , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
5.
Cell Chem Biol ; 25(8): 1006-1016.e8, 2018 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-29937406

RESUMO

Viral envelope proteins are required for productive viral entry and initiation of infection. Although the humoral immune system provides ample evidence for targeting envelope proteins as an antiviral strategy, there are few pharmacological interventions that have this mode of action. In contrast to classical antiviral targets such as viral proteases and polymerases, viral envelope proteins as a class do not have a well-conserved active site that can be rationally targeted with small molecules. We previously identified compounds that inhibit dengue virus by binding to its envelope protein, E. Here, we show that these small molecules inhibit dengue virus fusion and map the binding site of these compounds to a specific pocket on E. We further demonstrate inhibition of Zika, West Nile, and Japanese encephalitis viruses by these compounds, providing pharmacological evidence for the pocket as a target for developing broad-spectrum antivirals against multiple, mosquito-borne flavivirus pathogens.


Assuntos
Antivirais/química , Antivirais/farmacologia , Infecções por Flavivirus/tratamento farmacológico , Flavivirus/efeitos dos fármacos , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Linhagem Celular , Sequência Conservada , Vírus da Dengue/química , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/fisiologia , Descoberta de Drogas , Flavivirus/química , Flavivirus/fisiologia , Infecções por Flavivirus/metabolismo , Infecções por Flavivirus/virologia , Humanos , Simulação de Acoplamento Molecular , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas do Envelope Viral/química , Replicação Viral/efeitos dos fármacos , Zika virus/química , Zika virus/efeitos dos fármacos , Zika virus/fisiologia
6.
Antiviral Res ; 139: 171-179, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28034743

RESUMO

We report here on an approach targeting the host reactive cysteinome to identify inhibitors of host factors required for the infectious cycle of Flaviviruses and other viruses. We used two parallel cellular phenotypic screens to identify a series of covalent inhibitors, exemplified by QL-XII-47, that are active against dengue virus. We show that the compounds effectively block viral protein expression and that this inhibition is associated with repression of downstream processes of the infectious cycle, and thus significantly contributes to the potent antiviral activity of these compounds. We demonstrate that QL-XII-47's antiviral activity requires selective, covalent modification of a host target by showing that the compound's antiviral activity is recapitulated when cells are preincubated with QL-XII-47 and then washed prior to viral infection and by showing that QL-XII-47R, a non-reactive analog, lacks antiviral activity at concentrations more than 20-fold higher than QL-XII-47's IC90. QL-XII-47's inhibition of Zika virus, West Nile virus, hepatitis C virus, and poliovirus further suggests that it acts via a target mediating inhibition of these other medically relevant viruses. These results demonstrate the utility of screens targeting the host reactive cysteinome for rapid identification of compounds with potent antiviral activity.


Assuntos
Antivirais/química , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Descoberta de Drogas , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Antivirais/isolamento & purificação , Dengue/tratamento farmacológico , Dengue/virologia , Flavivirus/efeitos dos fármacos , Hepacivirus/efeitos dos fármacos , Humanos , Replicação Viral/efeitos dos fármacos , Vírus do Nilo Ocidental/efeitos dos fármacos , Zika virus/efeitos dos fármacos
7.
J Virol ; 89(11): 5862-75, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25787283

RESUMO

UNLABELLED: Japanese encephalitis virus (JEV) strains can be separated into 5 genotypes (g1 to g5) based on sequence similarity. JEV g5 strains have been rarely isolated and are poorly characterized. We report here the full characterization of a g5 virus generated using a cDNA-based technology and its comparison with a widely studied g3 strain. We did not observe any major differences between those viruses when their infectious cycles were studied in various cell lines in vitro. Interestingly, the JEV g5 strain was highly pathogenic when inoculated to BALB/c mice, which are known to be largely resistant to JEV g3 infection. The study of chimeric viruses between JEV g3 and g5 showed that there was a poor viral clearance of viruses that express JEV g5 structural proteins in BALB/c mice blood, which correlated with viral invasion of the central nervous system and encephalitis. In addition, using an in vitro model of the blood-brain barrier, we were able to show that JEV g5 does not have an enhanced capacity for entering the central nervous system, compared to JEV g3. Overall, in addition to providing a first characterization of the understudied JEV g5, our work highlights the importance of sustaining an early viremia in the development of JEV encephalitis. IMPORTANCE: Genotype 5 viruses are genetically and serologically distinct from other JEV genotypes and can been associated with human encephalitis, which warrants the need for their characterization. In this study, we characterized the in vitro and in vivo properties of a JEV g5 strain and showed that it was more neuropathogenic in a mouse model than a well-characterized JEV g3 strain. The enhanced virulence of JEV g5 was associated with poor viral clearance but not with enhanced crossing of the blood-brain barrier, thus providing new insights into JEV pathogenesis.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/classificação , Vírus da Encefalite Japonesa (Espécie)/patogenicidade , Encefalite Japonesa/virologia , Proteínas Estruturais Virais/metabolismo , Fatores de Virulência/metabolismo , Animais , Barreira Hematoencefálica/fisiologia , Barreira Hematoencefálica/virologia , Vírus da Encefalite Japonesa (Espécie)/isolamento & purificação , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Encefalite Japonesa/patologia , Feminino , Genótipo , Camundongos Endogâmicos BALB C , Proteínas Estruturais Virais/genética , Virulência , Fatores de Virulência/genética , Replicação Viral
8.
J Virol ; 87(13): 7367-81, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23616652

RESUMO

In this study, we characterized the antiviral mechanism of action of AZD0530 and dasatinib, two pharmacological inhibitors of host kinases, that also inhibit dengue virus (DV) infection. Using Northern blot and reporter replicon assays, we demonstrated that both small molecules inhibit the DV2 infectious cycle at the step of steady-state RNA replication. In order to identify the cellular target of AZD0530 and dasatinib mediating this anti-DV2 activity, we examined the effects of RNA interference (RNAi)-mediated depletion of the major kinases known to be inhibited by these small molecules. We determined that Fyn kinase, a target of both AZD0530 and dasatinib, is involved in DV2 RNA replication and is probably a major mediator of the anti-DV activity of these compounds. Furthermore, serial passaging of DV2 in the presence of dasatinib led to the identification of a mutation in the transmembrane domain 3 of the NS4B protein that overcomes the inhibition of RNA replication by AZD0530, dasatinib, and Fyn RNAi. Although we observed that dasatinib also inhibits DV2 particle assembly and/or secretion, this activity does not appear to be mediated by Src-family kinases. Together, our results suggest that AZD0530 and dasatinib inhibit DV at the step of viral RNA replication and demonstrate a critical role for Fyn kinase in this viral process. The antiviral activity of these compounds in vitro makes them useful pharmacological tools to validate Fyn or other host kinases as anti-DV targets in vivo.


Assuntos
Benzodioxóis/farmacologia , Vírus da Dengue/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Pirimidinas/farmacologia , Quinazolinas/farmacologia , RNA Viral/biossíntese , Tiazóis/farmacologia , Replicação Viral/efeitos dos fármacos , Anticorpos Monoclonais/imunologia , Northern Blotting , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Primers do DNA/genética , Dasatinibe , Vírus da Dengue/genética , Dimetil Sulfóxido , Células HEK293 , Humanos , Ácido Micofenólico , Reação em Cadeia da Polimerase , Interferência de RNA , Replicon/genética , Replicação Viral/fisiologia
9.
J Virol ; 86(13): 7072-83, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22532681

RESUMO

The dengue virus (DV) envelope (E) protein is important in mediating viral entry and assembly of progeny virus during cellular infection. Domains I and III (DI and DIII, respectively) of the DV E protein are connected by a highly conserved but poorly ordered region, the DI/DIII linker. Although the flexibility of the DI/DIII linker is thought to be important for accommodating the structural rearrangements undergone by the E protein during viral entry, the function of the linker in the DV infectious cycle is not well understood. In this study, we performed site-directed mutagenesis on conserved residues in the DI/DIII linker of the DV2 E protein and showed that the resulting mutations had little or no effect on the entry process but greatly affected virus assembly. Biochemical fractionation and immunofluorescence microscopy experiments performed on infectious virus as well as in a virus-like particle (VLP) system indicate that the DI/DIII linker mutants express the DV structural proteins at the sites of particle assembly near the ER but fail to form infectious particles. This defect is not due to disruption of E's interaction with prM and pr in immature and mature virions, respectively. Serial passaging of the DV2 mutant E-Y299F led to the identification of a mutation in the membrane-proximal stem region of E that fully compensates for the assembly defect of this DI/DIII linker mutant. Together, our results suggest a critical and previously unidentified role for the E protein DI/DIII linker region during the DV2 assembly process.


Assuntos
Vírus da Dengue/fisiologia , Mutagênese Sítio-Dirigida , Proteínas do Envelope Viral/metabolismo , Montagem de Vírus , Sequência de Aminoácidos , Linhagem Celular , Vírus da Dengue/genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas do Envelope Viral/genética , Internalização do Vírus
10.
Virology ; 384(1): 179-91, 2009 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19059620

RESUMO

Cucumber Mosaic Virus (CMV) is a plant infecting tripartite positive-strand RNA virus. In addition to three genomic and two known subgenomic RNAs, CMV strains of subgroup II (e.g. Q-CMV), but not subgroup I (e.g. Fny-CMV), produce and package a redundant RNA5 encompassing the 3' 304-307 nucleotides of RNAs 2 and 3. The mechanism regulating RNA5 production and its role in CMV life cycle is unknown. In this study, transient expression of Q2 or Q3 by agroinfiltration into Nicotiana benthamiana plants resulted in efficient accumulation of RNA5 suggesting that its production is independent of CMV replication. Deletion and point mutations engineered into a highly conserved region (Box1) adjacent to the 5' end of RNA5 identified sequences required for its efficient production. An experimental system, involving a chimera of Q3 (Q3B3) characterized by having a 3' tRNA-like structure (3'TLS) from Brome mosaic virus (BMV) and RNA5 defective variants of Q1 (Q1Delta), Q2 (Q2Delta) and Q3B3 (Q3DeltaB3), was used to evaluate in vivo the contribution of RNA5 in promoting RNA recombination. Generation of precise homologous recombinants was strictly dependent on sequence identity. When both parental RNAs carried the Box1, recombination occurred preferentially within the Box1. In contrast, generation of non-homologous recombinants occurred only when Q1 and Q2 were competent to produce RNA5. A mechanistic model explaining the functional role played by the RNA5 in generating CMV recombinants was presented.


Assuntos
Cucumovirus/genética , RNA Viral/genética , Recombinação Genética , Sequência Conservada , Cucumis sativus/virologia , DNA Complementar/genética , DNA Viral/genética , Regulação Viral da Expressão Gênica , Genoma Viral , Doenças das Plantas/virologia , Plasmídeos , Rhizobium/genética , Transcrição Gênica , Replicação Viral
11.
Virology ; 331(1): 117-27, 2005 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-15582658

RESUMO

In order to better understand the role of recombination in creating the diversity of viral genomes that is acted on by selection, we have studied in detail the population of recombinant RNA3 molecules occurring in tobacco plants coinfected with wild-type strains of cucumber mosaic virus (CMV) and tomato aspermy virus (TAV) under conditions of minimal selection pressure. Recombinant RNA3s were observed in 9.6% of the samples. Precise homologous recombination predominated since it was observed at 28 different sites, primarily in six hot spots. Imprecise homologous recombination was observed at two sites, particularly within a GU repeat in the 5' noncoding region. Seven of the eight aberrant homologous recombination sites observed were clustered in the 3' noncoding region. These results have implications on the role of recombination in host adaptation and virus evolution. They also provide essential baseline information for understanding the potential epidemiological impact of recombination in transgenic plants expressing viral sequences.


Assuntos
Cucumovirus/genética , Doenças das Plantas/genética , Doenças das Plantas/virologia , RNA Viral/genética , RNA/genética , Sequência de Bases , Cucumis sativus/virologia , Variação Genética , Solanum lycopersicum/virologia , Dados de Sequência Molecular , Recombinação Genética , Homologia de Sequência do Ácido Nucleico , Nicotiana/genética , Nicotiana/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA