Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Biosci Rep ; 43(5)2023 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-37078499

RESUMO

Retinoic acid inducible gene (RIG)-I-like receptors (RLRs), including RIG-I, melanoma differentiation associated-5 (MDA5), and laboratory of genetics and physiology 2 (LGP2), play pivotal roles in viral RNA sensing to initiate antiviral interferon (IFN) responses. We previously reported that an RNA-silencing regulator, transactivation response RNA-binding protein (TRBP), up-regulates MDA5/LGP2-mediated IFN responses through interaction with LGP2. Here, we aimed to investigate the mechanism underlying the TRBP-mediated up-regulation of IFN response. Data indicated that phosphomimetic TRBP showed a modest effect, whereas the nonphosphorylated form exhibited hyperactivity in enhancing Cardiovirus-triggered IFN responses. These results suggest that encephalomyocarditis virus (EMCV) attenuates the TRBP-mediated IFN response via TRBP phosphorylation, since EMCV infection activates the kinase responsible for TRBP phosphorylation for virus replication. Furthermore, we found that TRBP-mediated up-regulation of IFN response required the ATP hydrolysis and RNA binding of LGP2. TRBP enhanced RNA-dependent ATP hydrolysis by LGP2 but not that by RIG-I or MDA5. Nonphosphorylated TRBP exhibited higher levels of activity than phosphomimetic TRBP did, suggesting its possible involvement in the mechanism underlying the up-regulation of IFN response. TRBP activated the ATP hydrolysis of LGP2 and RIG-I, but not that of MDA5, in the absence of RNA. Collectively, we showed that TRBP differentially regulated RLR-mediated ATP hydrolysis. Further elucidation of the mechanism underlying the regulation of ATP hydrolysis leading to IFN response and self- and non-self-RNA discrimination could advance the development of effective therapeutic agents against autoimmune diseases.


Assuntos
Vírus da Encefalomiocardite , RNA Helicases , RNA Helicases/genética , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/metabolismo , Ativação Transcricional , Hidrólise , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , RNA Viral/genética , Trifosfato de Adenosina , Imunidade Inata
2.
J Virol ; 95(6)2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-33328314

RESUMO

Type I interferon (IFN)-mediated antiviral responses are critical for modulating host-virus responses, and indeed, viruses have evolved strategies to antagonize this pathway. Encephalomyocarditis virus (EMCV) is an important zoonotic pathogen, which causes myocarditis, encephalitis, neurological disease, reproductive disorders, and diabetes in pigs. This study aims to understand how EMCV interacts with the IFN pathway. EMCV circumvents the type I IFN response by expressing proteins that antagonize cellular innate immunity. Here, we show that EMCV VP2 is a negative regulator of the IFN-ß pathway. This occurs via the degradation of the MDA5-mediated cytoplasmic double-stranded RNA (dsRNA) antiviral sensing RIG-I-like receptor (RLR) pathway. We show that structural protein VP2 of EMCV interacts with MDA5, MAVS, and TBK1 through its C terminus. In addition, we found that EMCV VP2 could significantly degrade RLRs by the proteasomal and lysosomal pathways. For the first time, EMCV VP2 was shown to play an important role in EMCV evasion of the type I IFN signaling pathway. This study expands our understanding that EMCV utilizes its capsid protein VP2 to evade the host antiviral response.IMPORTANCE Encephalomyocarditis virus is an important pathogen that can cause encephalitis, myocarditis, neurological diseases, and reproductive disorders. It also causes huge economic losses for the swine industry worldwide. Innate immunity plays an important role in defending the host from pathogen infection. Understanding pathogen microorganisms evading the host immune system is of great importance. Currently, whether EMCV evades cytosolic RNA sensing and signaling is still poorly understood. In the present study, we found that viral protein VP2 antagonized the RLR signaling pathway by degrading MDA5, MAVS, and TBK1 protein expression to facilitate viral replication in HEK293 cells. The findings in this study identify a new mechanism for EMCV evading the host's innate immune response, which provide new insights into the virus-host interaction and help develop new antiviral approaches against EMCV.


Assuntos
Proteínas do Capsídeo/metabolismo , Vírus da Encefalomiocardite/fisiologia , Interferon beta/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/virologia , Proteína DEAD-box 58/antagonistas & inibidores , Proteína DEAD-box 58/metabolismo , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/metabolismo , Células HEK293 , Humanos , Evasão da Resposta Imune , Imunidade Inata , Interferon Tipo I/metabolismo , Helicase IFIH1 Induzida por Interferon/metabolismo , Mutação , Domínios e Motivos de Interação entre Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/metabolismo , Replicação Viral
3.
Nat Struct Mol Biol ; 27(12): 1095-1104, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32958947

RESUMO

Viruses use internal ribosome entry sites (IRES) to hijack host ribosomes and promote cap-independent translation. Although they are well-studied in bulk, the dynamics of IRES-mediated translation remain unexplored at the single-molecule level. Here, we developed a bicistronic biosensor encoding distinct repeat epitopes in two open reading frames (ORFs), one translated from the 5' cap, and the other from the encephalomyocarditis virus IRES. When combined with a pair of complementary probes that bind the epitopes cotranslationally, the biosensor lights up in different colors depending on which ORF is translated. Using the sensor together with single-molecule tracking and computational modeling, we measured the kinetics of cap-dependent versus IRES-mediated translation in living human cells. We show that bursts of IRES translation are shorter and rarer than bursts of cap translation, although the situation reverses upon stress. Collectively, our data support a model for translational regulation primarily driven by transitions between translationally active and inactive RNA states.


Assuntos
Vírus da Encefalomiocardite/genética , Células Epiteliais/metabolismo , Sítios Internos de Entrada Ribossomal , Biossíntese de Proteínas , Capuzes de RNA/genética , Pareamento de Bases , Técnicas Biossensoriais , Linhagem Celular Tumoral , Vírus da Encefalomiocardite/metabolismo , Células Epiteliais/virologia , Epitopos/química , Epitopos/genética , Epitopos/metabolismo , Interações Hospedeiro-Patógeno/genética , Humanos , Sequências Repetidas Invertidas , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Cinesinas/genética , Cinesinas/metabolismo , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Conformação de Ácido Nucleico , Fases de Leitura Aberta , Capuzes de RNA/química , Capuzes de RNA/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Ribossomos/genética , Ribossomos/metabolismo , Imagem Individual de Molécula/métodos
4.
Bol. latinoam. Caribe plantas med. aromát ; 19(6): 542-554, 2020. ilus, tab
Artigo em Inglês | LILACS | ID: biblio-1284288

RESUMO

The enrichment of therapeutic protein production yield in mammalian cell cultures by modulating mRNA stability is a fairly new strategy in biotechnological applications. Here, we describe the application of 3'-untranslated region (3'UTR) from RNA viral genome to modulate mRNA stability.The data obtained showed that the use of the 3 'UTR sequence of the encephalomyocarditis virus (EMCV 3'UTR) downstream of the target gene was not able to significantly modulate the free energy density indicators of the RNA. However, the sequence influenced the stability of the mRNA (and, therefore, the amount of protein production) in a cell type and time-dependent manner, indicating a central role of mRNA-stabilizing binding sites/cellular factors in this process. Our data might be of interest for the biotechnology community to improve recombinant protein production in mammalian cell cultures and RNA-based therapy/vaccination approaches.


El enriquecimiento de la producción terapéutica de proteínas en cultivos de células de mamíferos mediante la modulación de la estabilidad del ARNm es una estrategia nueva en aplicaciones biotecnológicas. Se describe la aplicación de la región 3'-no traducida (3'UTR) del genoma viral ARN para modular la estabilidad del ARNm. Los datos obtenidos mostraron que el uso de la secuencia 3'UTR del virus de la encefalomiocarditis (EMCV 3'UTR) aguas abajo del gen objetivo no pudo modular significativamente los indicadores de densidad de energía libre del ARN. Sin embargo, la secuencia influyó en la estabilidad del ARNm (y, por lo tanto, en la cantidad de producción de proteínas) dependiente de la célula y del tiempo, lo que indica un papel central de los sitios de unión estabilizadores de ARNm/factores celulares en este proceso. Nuestros datos podrían ser de interés para la comunidad biotecnológica para mejorar la producción de proteínas recombinantes en cultivos de células de mamíferos y en enfoques de terapia/vacunación basados en ARN.


Assuntos
Produtos Biológicos , Proteínas Recombinantes/biossíntese , Regiões não Traduzidas , Proteínas de Fluorescência Verde/metabolismo , Vírus da Encefalomiocardite/metabolismo , Biotecnologia , Genoma Viral , Técnicas de Cultura de Células , Estabilidade de RNA , Vírus da Encefalomiocardite/genética
5.
J Virol ; 94(1)2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31597774

RESUMO

Hepatitis C virus (HCV) nonstructural protein 2 (NS2) is a multifunctional protein implicated in both HCV RNA replication and virus particle assembly. NS2-encoded cysteine protease is responsible for autoprocessing of NS2-NS3 precursor, an essential step in HCV RNA replication. NS2 also promotes HCV particle assembly by recruiting envelope protein 2 (E2) to the virus assembly sites located at the detergent-resistant membranes (DRM). However, the fundamental mechanism regulating multiple functions of NS2 remains unclear. In this study, we discovered that NS2 is palmitoylated at the position 113 cysteine residue (NS2/C113) when expressed by itself in cells and during infectious-HCV replication. Blocking NS2 palmitoylation by introducing an NS2/C113S mutation reduced NS2-NS3 autoprocessing and impaired HCV RNA replication. Replication of the NS2/C113S mutant was restored by inserting an encephalomyocarditis virus (EMCV) internal ribosome entry site (IRES) between NS2 and NS3 to separate the two proteins independently of NS2-mediated autoprocessing. These results suggest that NS2 palmitoylation is critical for HCV RNA replication by promoting NS2-NS3 autoprocessing. The NS2/C113S mutation also impaired infectious-HCV assembly, DRM localization of NS2 and E2, and colocalization of NS2 with Core and endoplasmic reticulum lipid raft-associated protein 2 (Erlin-2). In conclusion, our study revealed that two major functions of NS2 involved in HCV RNA replication and virus assembly, i.e., NS2-NS3 autoprocessing and E2 recruitment to the DRM, are regulated by palmitoylation at NS2/C113. Since S-palmitoylation is reversible, NS2 palmitoylation likely allows NS2 to fine tune both HCV RNA replication and infectious-particle assembly.IMPORTANCE Chronic infection with hepatitis C virus (HCV) is a major cause of severe liver diseases responsible for nearly 400,000 deaths per year. HCV NS2 protein is a multifunctional regulator of HCV replication involved in both viral-genome replication and infectious-virus assembly. However, the underlying mechanism that enables the protein to participate in multiple steps of HCV replication remains unknown. In this study, we discovered that NS2 palmitoylation is the master regulator of its multiple functions, including NS2-mediated self-cleavage and HCV envelope protein recruitment to the virus assembly sites, which in turn promote HCV RNA replication and infectious-particle assembly, respectively. This newly revealed information suggests that NS2 palmitoylation could serve as a promising target to inhibit both HCV RNA replication and virus assembly, representing a new avenue for host-targeting strategies against HCV infection.


Assuntos
Hepacivirus/metabolismo , Interações Hospedeiro-Patógeno/genética , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas não Estruturais Virais/metabolismo , Sequência de Aminoácidos , Linhagem Celular Tumoral , Cisteína/química , Cisteína/metabolismo , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/metabolismo , Células HEK293 , Hepacivirus/genética , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , Lipoilação , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Moleculares , Mutação , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Precursores de Proteínas/química , Precursores de Proteínas/genética , Transporte Proteico , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Carga Viral , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Replicação Viral
6.
mBio ; 10(4)2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31409686

RESUMO

Encephalomyocarditis virus (EMCV) is an animal pathogen and an important model organism, whose receptor requirements are poorly understood. Here, we employed a genome-wide haploid genetic screen to identify novel EMCV host factors. In addition to the previously described picornavirus receptors sialic acid and glycosaminoglycans, this screen unveiled important new host factors for EMCV. These factors include components of the fibroblast growth factor (FGF) signaling pathway, such as the potential receptors FGFR1 and ADAM9, a cell-surface metalloproteinase. By employing various knockout cells, we confirmed the importance of the identified host factors for EMCV infection. The largest reduction in infection efficiency was observed in cells lacking ADAM9. Pharmacological inhibition of the metalloproteinase activity of ADAM9 did not affect virus infection. Moreover, reconstitution of inactive ADAM9 in knockout cells restored susceptibility to EMCV, pointing to a proteinase-independent role of ADAM9 in mediating EMCV infection. Using neutralization assays with ADAM9-specific antiserum and soluble receptor proteins, we provided evidence for a role of ADAM9 in EMCV entry. Finally, binding assays showed that ADAM9 facilitates attachment of EMCV to the cell surface. Together, our findings reveal a role for ADAM9 as a novel receptor or cofactor for EMCV.IMPORTANCE EMCV is an animal pathogen that causes acute viral infections, usually myocarditis or encephalitis. It is thought to circulate mainly among rodents, from which it is occasionally transmitted to other animal species, including humans. EMCV causes fatal outbreaks of myocarditis and encephalitis in pig farms and zoos, making it an important veterinary pathogen. Although EMCV has been widely used as a model to study mechanisms of viral disease in mice, little is known about its entry mechanism. Here, we employ a haploid genetic screen for EMCV host factors and identify an essential role for ADAM9 in EMCV entry.


Assuntos
Proteínas ADAM/metabolismo , Infecções por Cardiovirus/virologia , Vírus da Encefalomiocardite/fisiologia , Proteínas de Membrana/metabolismo , Internalização do Vírus , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/genética , Animais , Infecções por Cardiovirus/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Vírus da Encefalomiocardite/metabolismo , Técnicas de Inativação de Genes , Genoma Humano/genética , Humanos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Camundongos , Ligação Viral , Replicação Viral
7.
Virology ; 487: 19-26, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26492198

RESUMO

Cardiovirus Leader proteins (LX) inhibit cellular nucleocytoplasmic trafficking by directing host kinases to phosphorylate Phe/Gly-containing nuclear pore proteins (Nups). Resolution of the Mengovirus LM structure bound to Ran GTPase, suggested this complex would further recruit specific exportins (karyopherins), which in turn mediate kinase selection. Pull-down experiments and recombinant complex reconstitution now confirm that Crm1 and CAS exportins form stable dimeric complexes with encephalomyocarditis virus LE, and also larger complexes with LE:Ran. shRNA knockdown studies support this idea. Similar activities could be demonstrated for recombinant LS and LT from Theiloviruses. When mutations were introduced to alter the LE zinc finger domain, acidic domain, or dual phosphorylation sites, there was reduced exportin selection. These regions are not involved in Ran interactions, so the Ran and Crm1 binding sites on LE must be non-overlapping. The involvement of exportins in this mechanism is important to viral replication and the observation of trafficking inhibition by LE.


Assuntos
Transporte Ativo do Núcleo Celular/genética , Cardiovirus/metabolismo , Carioferinas/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteína ran de Ligação ao GTP/metabolismo , Sítios de Ligação/genética , Linhagem Celular Tumoral , Vírus da Encefalomiocardite/metabolismo , Células HeLa , Humanos , Carioferinas/genética , Mengovirus/metabolismo , Fosforilação , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno , Receptores Citoplasmáticos e Nucleares/genética , Theilovirus/metabolismo , Replicação Viral/genética , Proteína ran de Ligação ao GTP/genética , Proteína Exportina 1
8.
J Biol Chem ; 290(32): 19681-96, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26055709

RESUMO

2',5'-Oligoadenylate synthetase (OAS) enzymes and RNase-L constitute a major effector arm of interferon (IFN)-mediated antiviral defense. OAS produces a unique oligonucleotide second messenger, 2',5'-oligoadenylate (2-5A), that binds and activates RNase-L. This pathway is down-regulated by virus- and host-encoded enzymes that degrade 2-5A. Phosphodiesterase 12 (PDE12) was the first cellular 2-5A- degrading enzyme to be purified and described at a molecular level. Inhibition of PDE12 may up-regulate the OAS/RNase-L pathway in response to viral infection resulting in increased resistance to a variety of viral pathogens. We generated a PDE12-null cell line, HeLaΔPDE12, using transcription activator-like effector nuclease-mediated gene inactivation. This cell line has increased 2-5A levels in response to IFN and poly(I-C), a double-stranded RNA mimic compared with the parental cell line. Moreover, HeLaΔPDE12 cells were resistant to viral pathogens, including encephalomyocarditis virus, human rhinovirus, and respiratory syncytial virus. Based on these results, we used DNA-encoded chemical library screening to identify starting points for inhibitor lead optimization. Compounds derived from this effort raise 2-5A levels and exhibit antiviral activity comparable with the effects observed with PDE12 gene inactivation. The crystal structure of PDE12 complexed with an inhibitor was solved providing insights into the structure-activity relationships of inhibitor potency and selectivity.


Assuntos
2',5'-Oligoadenilato Sintetase/imunologia , Antivirais/farmacologia , Endorribonucleases/imunologia , Exorribonucleases/química , Imunidade Inata , Bibliotecas de Moléculas Pequenas/farmacologia , 2',5'-Oligoadenilato Sintetase/genética , Nucleotídeos de Adenina/imunologia , Nucleotídeos de Adenina/metabolismo , Antivirais/síntese química , Cristalografia por Raios X , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/metabolismo , Endorribonucleases/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Exorribonucleases/antagonistas & inibidores , Exorribonucleases/genética , Exorribonucleases/imunologia , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Células HeLa , Humanos , Interferon-alfa/farmacologia , Modelos Moleculares , Oligorribonucleotídeos/imunologia , Oligorribonucleotídeos/metabolismo , Poli I-C/farmacologia , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/metabolismo , Rhinovirus/genética , Rhinovirus/metabolismo , Transdução de Sinais , Bibliotecas de Moléculas Pequenas/síntese química , Relação Estrutura-Atividade
9.
J Biol Chem ; 289(46): 31960-31971, 2014 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-25258322

RESUMO

The genomic RNA of encephalomyocarditis virus (EMCV) encodes a single polyprotein, and the primary scission of the polyprotein occurs between nonstructural proteins 2A and 2B by an unknown mechanism. To gain insight into the mechanism of 2A-2B processing, we first translated the 2A-2B region in vitro with eukaryotic and prokaryotic translation systems. The 2A-2B processing occurred only in the eukaryotic systems, not in the prokaryotic systems, and the unprocessed 2A-2B protein synthesized by a prokaryotic system remained uncleaved when incubated with a eukaryotic cell extract. These results suggest that 2A-2B processing is a eukaryote-specific, co-translational event. To define the translation factors required for 2A-2B processing, we constituted a protein synthesis system with eukaryotic elongation factors 1 and 2, eukaryotic release factors 1 and 3 (eRF1 and eRF3), aminoacyl-tRNA synthetases, tRNAs, ribosome subunits, and a plasmid template that included the hepatitis C virus internal ribosome entry site. We successfully reproduced 2A-2B processing in the reconstituted system even without eRFs. Our results indicate that this unusual event occurs in the elongation phase of translation.


Assuntos
Vírus da Encefalomiocardite/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator 1 de Elongação de Peptídeos/metabolismo , Biossíntese de Proteínas , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/metabolismo , Alanina/genética , Aminoacil-tRNA Sintetases/metabolismo , Sítios de Ligação , Sistema Livre de Células , DNA Complementar/metabolismo , Deleção de Genes , Regulação da Expressão Gênica , Células HeLa , Hepacivirus/metabolismo , Humanos , Mutação , Fases de Leitura Aberta , Plasmídeos/metabolismo , Dobramento de Proteína , Ribossomos/química
10.
J Virol ; 88(19): 11091-107, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25008939

RESUMO

UNLABELLED: Few drugs targeting picornaviruses are available, making the discovery of antivirals a high priority. Here, we identified and characterized three compounds from a library of kinase inhibitors that block replication of poliovirus, coxsackievirus B3, and encephalomyocarditis virus. Using an in vitro translation-replication system, we showed that these drugs inhibit different stages of the poliovirus life cycle. A4(1) inhibited both the formation and functioning of the replication complexes, while E5(1) and E7(2) were most effective during the formation but not the functioning step. Neither of the compounds significantly inhibited VPg uridylylation. Poliovirus resistant to E7(2) had a G5318A mutation in the 3A protein. This mutation was previously found to confer resistance to enviroxime-like compounds, which target a phosphatidylinositol 4-kinase IIIß (PI4KIIIß)-dependent step in viral replication. Analysis of host protein recruitment showed that E7(2) reduced the amount of GBF1 on the replication complexes; however, the level of PI4KIIIß remained intact. E7(2) as well as another enviroxime-like compound, GW5074, interfered with viral polyprotein processing affecting both 3C- and 2A-dependent cleavages, and the resistant G5318A mutation partially rescued this defect. Moreover, E7(2) induced abnormal recruitment to membranes of the viral proteins; thus, enviroxime-like compounds likely severely compromise the interaction of the viral polyprotein with membranes. A4(1) demonstrated partial protection from paralysis in a murine model of poliomyelitis. Multiple attempts to isolate resistant mutants in the presence of A4(1) or E5(1) were unsuccessful, showing that effective broad-spectrum antivirals could be developed on the basis of these compounds. IMPORTANCE: Diverse picornaviruses can trigger multiple human maladies, yet currently, only hepatitis A virus and poliovirus can be controlled with vaccination. The development of antipicornavirus therapeutics is also facing significant difficulties because these viruses readily generate resistance to compounds targeting either viral or cellular factors. Here, we describe three novel compounds that effectively block replication of distantly related picornaviruses with minimal toxicity to cells. The compounds prevent viral RNA replication after the synthesis of the uridylylated VPg primer. Importantly, two of the inhibitors are strongly refractory to the emergence of resistant mutants, making them promising candidates for further broad-spectrum therapeutic development. Evaluation of one of the compounds in an in vivo model of poliomyelitis demonstrated partial protection from the onset of paralysis.


Assuntos
Antivirais/farmacologia , Poliomielite/tratamento farmacológico , Poliovirus/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas Virais/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , 1-Fosfatidilinositol 4-Quinase/antagonistas & inibidores , 1-Fosfatidilinositol 4-Quinase/genética , 1-Fosfatidilinositol 4-Quinase/metabolismo , Animais , Antivirais/química , Sistema Livre de Células , Modelos Animais de Doenças , Vírus da Encefalomiocardite/efeitos dos fármacos , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/metabolismo , Enterovirus Humano B/efeitos dos fármacos , Enterovirus Humano B/genética , Enterovirus Humano B/metabolismo , Regulação Viral da Expressão Gênica , Células HeLa , Humanos , Camundongos , Mutação , Poliomielite/virologia , Poliovirus/genética , Poliovirus/crescimento & desenvolvimento , Poliproteínas/antagonistas & inibidores , Poliproteínas/genética , Poliproteínas/metabolismo , Transdução de Sinais , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade , Proteínas Virais/genética , Proteínas Virais/metabolismo
11.
Elife ; 3: e01535, 2014 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-24550253

RESUMO

The RIG-I-like receptors RIG-I, LGP2, and MDA5 initiate an antiviral response that includes production of type I interferons (IFNs). The nature of the RNAs that trigger MDA5 activation in infected cells remains unclear. Here, we purify and characterise LGP2/RNA complexes from cells infected with encephalomyocarditis virus (EMCV), a picornavirus detected by MDA5 and LGP2 but not RIG-I. We show that those complexes contain RNA that is highly enriched for MDA5-stimulatory activity and for a specific sequence corresponding to the L region of the EMCV antisense RNA. Synthesis of this sequence by in vitro transcription is sufficient to generate an MDA5 stimulatory RNA. Conversely, genomic deletion of the L region in EMCV generates viruses that are less potent at stimulating MDA5-dependent IFN production. Thus, the L region antisense RNA of EMCV is a key determinant of innate immunity to the virus and represents an RNA that activates MDA5 in virally-infected cells. DOI: http://dx.doi.org/10.7554/eLife.01535.001.


Assuntos
RNA Helicases DEAD-box/metabolismo , Vírus da Encefalomiocardite/metabolismo , RNA Helicases/metabolismo , RNA Antissenso/metabolismo , RNA Viral/metabolismo , Animais , Antivirais/farmacologia , Chlorocebus aethiops , RNA Helicases DEAD-box/genética , Vírus da Encefalomiocardite/efeitos dos fármacos , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/imunologia , Regulação Viral da Expressão Gênica , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Helicase IFIH1 Induzida por Interferon , Interferons/genética , Interferons/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , RNA Helicases/genética , RNA Antissenso/genética , RNA Viral/genética , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Transdução de Sinais , Transfecção , Células Vero , Replicação Viral
12.
PLoS Pathog ; 8(8): e1002857, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22916014

RESUMO

Nod-like receptors (NLRs) comprise a large family of intracellular pattern- recognition receptors. Members of the NLR family assemble into large multiprotein complexes, termed the inflammasomes. The NLR family, pyrin domain-containing 3 (NLRP3) is triggered by a diverse set of molecules and signals, and forms the NLRP3 inflammasome. Recent studies have indicated that both DNA and RNA viruses stimulate the NLRP3 inflammasome, leading to the secretion of interleukin 1 beta (IL-1ß) and IL-18 following the activation of caspase-1. We previously demonstrated that the proton-selective ion channel M2 protein of influenza virus activates the NLRP3 inflammasome. However, the precise mechanism by which NLRP3 recognizes viral infections remains to be defined. Here, we demonstrate that encephalomyocarditis virus (EMCV), a positive strand RNA virus of the family Picornaviridae, activates the NLRP3 inflammasome in mouse dendritic cells and macrophages. Although transfection with RNA from EMCV virions or EMCV-infected cells induced robust expression of type I interferons in macrophages, it failed to stimulate secretion of IL-1ß. Instead, the EMCV viroporin 2B was sufficient to cause inflammasome activation in lipopolysaccharide-primed macrophages. While cells untransfected or transfected with the gene encoding the EMCV non-structural protein 2A or 2C expressed NLRP3 uniformly throughout the cytoplasm, NLRP3 was redistributed to the perinuclear space in cells transfected with the gene encoding the EMCV 2B or influenza virus M2 protein. 2B proteins of other picornaviruses, poliovirus and enterovirus 71, also caused the NLRP3 redistribution. Elevation of the intracellular Ca(2+) level, but not mitochondrial reactive oxygen species and lysosomal cathepsin B, was important in EMCV-induced NLRP3 inflammasome activation. Chelation of extracellular Ca(2+) did not reduce virus-induced IL-1ß secretion. These results indicate that EMCV activates the NLRP3 inflammasome by stimulating Ca(2+) flux from intracellular storages to the cytosol, and highlight the importance of viroporins, transmembrane pore-forming viral proteins, in virus-induced NLRP3 inflammasome activation.


Assuntos
Cálcio/metabolismo , Infecções por Cardiovirus/metabolismo , Proteínas de Transporte/metabolismo , Vírus da Encefalomiocardite/metabolismo , Inflamassomos/metabolismo , Proteínas Virais/metabolismo , Animais , Infecções por Cardiovirus/genética , Proteínas de Transporte/genética , Vírus da Encefalomiocardite/genética , Células HEK293 , Células HeLa , Humanos , Inflamassomos/genética , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR , RNA Viral/genética , RNA Viral/metabolismo , Proteínas Virais/genética , Vírion/genética , Vírion/metabolismo
13.
Int J Mol Sci ; 13(6): 7365-7374, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22837699

RESUMO

The single-stranded RNA encephalomyocarditis virus (EMCV) can replicate in the central nervous system (CNS) and lead to prominent brain lesions in the stratum pyramidale hippocampus and the stratum granulosum cerebelli. Activated microglia cells infected by EMCV produce a massive burst of reactive oxygen species (ROS) via NADPH oxidase 2 (NOX2) activation, leading to neuronal death. Balancing this effect is mechanisms by which ROS are eliminated from the CNS. Cellular prion protein (PrP(C)) plays an important antioxidant role and contributes to cellular defense against EMCV infection. This review introduces recent knowledge on brain injury induced by EMCV infection via ROS generation as well as the involvement of various mediators and regulators in the pathogenesis.


Assuntos
Infecções por Cardiovirus/metabolismo , Vírus da Encefalomiocardite/metabolismo , Hipocampo/metabolismo , Glicoproteínas de Membrana/metabolismo , Microglia/metabolismo , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Infecções por Cardiovirus/patologia , Hipocampo/patologia , Hipocampo/virologia , Humanos , Microglia/patologia , Microglia/virologia , NADPH Oxidase 2 , Proteínas PrPC/metabolismo
14.
RNA ; 17(6): 1120-31, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21518806

RESUMO

Polypyrimidine tract binding protein (PTB) is an RNA-binding protein with four RNA-binding domains (RBDs). It is a major regulator of alternative splicing and also stimulates translation initiation at picornavirus IRESs (internal ribosome entry sites). The sites of interaction of each RBD with two picornaviral IRESs have previously been mapped. To establish which RBD-IRES interactions are essential for IRES activation, point mutations were introduced into the RNA-binding surface of each RBD. Three such mutations were sufficient to inactivate RNA-binding by any one RBD, but the sites of the other three RBD-IRES interactions remained unperturbed. Poliovirus IRES activation was abrogated by inactivation of RBD1, 2, or 4, but the RBD3-IRES interaction was superfluous. Stimulation of the encephalomyocarditis virus IRES was reduced by inactivation of RBD1, 3, or 4, and abrogated by mutation of RBD2, or both RBDs 3 and 4. Surprisingly, therefore, the binding of PTB in its normal orientation does not guarantee IRES activation; three native RBDs are sufficient for correct binding but not for activation if the missing RBD-IRES interaction is critical.


Assuntos
Picornaviridae/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Biossíntese de Proteínas , RNA Viral/química , RNA Viral/metabolismo , Sítios de Ligação , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/metabolismo , Conformação de Ácido Nucleico , Picornaviridae/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/química , Estrutura Terciária de Proteína , Ribossomos/genética , Ribossomos/metabolismo
16.
Virology ; 393(2): 311-8, 2009 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-19733381

RESUMO

RNA helicase-like receptors MDA-5 but not RIG-I has been shown to be essential for triggering innate immune responses against picornaviruses. However, virus-host co-evolution has selected for viruses capable of replicating despite host cells antiviral defences. In this report, we demonstrate that RIG-I is degraded during encephalomyocarditis virus (EMCV) infection. This effect is mediated by both the viral-encoded 3C protease and caspase proteinase. In addition, we show that RIG-I overexpression confers IFN-beta promoter activation during EMCV infection, in MDA-5 knockout (MDA-5(-/-)) mouse embryo fibroblasts. This induction is followed by a strong inhibition reflecting the ability of EMCV to disrupt RIG-I signalling. Taken together, our data strongly suggest that during evolution RIG-I has been involved for triggering innate immune response to picornavirus infections.


Assuntos
Infecções por Cardiovirus/imunologia , RNA Helicases DEAD-box/metabolismo , Vírus da Encefalomiocardite/imunologia , Imunidade Inata , Interferon beta/imunologia , Proteases Virais 3C , Animais , Infecções por Cardiovirus/virologia , Caspases/imunologia , Caspases/metabolismo , Cisteína Endopeptidases/imunologia , Cisteína Endopeptidases/metabolismo , Proteína DEAD-box 58 , RNA Helicases DEAD-box/imunologia , Vírus da Encefalomiocardite/metabolismo , Regulação da Expressão Gênica , Células HeLa , Humanos , Helicase IFIH1 Induzida por Interferon , Camundongos , Regiões Promotoras Genéticas , Proteínas Virais/imunologia , Proteínas Virais/metabolismo
17.
BMC Biotechnol ; 7: 85, 2007 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-18053131

RESUMO

BACKGROUND: Retroviral vectors are valuable tools for gene transfer. Particularly convenient are IRES-containing retroviral vectors expressing both the protein of interest and a marker protein from a single bicistronic mRNA. This coupled expression increases the relevance of tracking and/or selection of transduced cells based on the detection of a marker protein. pAP2 is a retroviral vector containing eGFP downstream of a modified IRES element of EMCV origin, and a CMV enhancer-promoter instead of the U3 region of the 5'LTR, which increases its efficiency in transient transfection. However, pAP2 contains a limited multicloning site (MCS) and shows weak eGFP expression, which previously led us to engineer an improved version, termed pPRIG, harboring: i) the wild-type ECMV IRES sequence, thereby restoring its full activity; ii) an optimized MCS flanked by T7 and SP6 sequences; and iii) a HA tag encoding sequence 5' of the MCS (pPRIG HAa/b/c). RESULTS: The convenience of pPRIG makes it a good basic vector to generate additional derivatives for an extended range of use. Here we present several novel pPRIG-based vectors (collectively referred to as PRIGs) in which : i) the HA tag sequence was inserted in the three reading frames 3' of the MCS (3'HA PRIGs); ii) a functional domain (ER, VP16 or KRAB) was inserted either 5' or 3' of the MCS (<< modular >> PRIGs); iii) eGFP was replaced by either eCFP, eYFP, mCherry or puro-R (<< single color/resistance >> PRIGs); and iv) mCherry, eYFP or eGFP was inserted 5' of the MCS of the IRES-eGFP, IRES-eCFP or IRES-Puro-R containing PRIGs, respectively (<< dual color/selection >> PRIGs). Additionally, some of these PRIGs were also constructed in a pMigR MSCV background which has been widely used in pluripotent cells. CONCLUSION: These novel vectors allow for straightforward detection of any expressed protein (3'HA PRIGs), for functional studies of chimeric proteins (<< modular >> PRIGs), for multiple transductions and fluorescence analyses of transduced cells (<< single color/resistance >> PRIGs), or for quantitative detection of studied proteins in independently identified/selected transduced cells (<< dual color/selection >> PRIGs). They maintain the original advantages of pPRIG and provide suitable tools for either transient or stable expression and functional studies in a large range of experimental settings.


Assuntos
Vírus da Encefalomiocardite/genética , Vetores Genéticos , Proteínas Luminescentes , Sequência de Bases , Clonagem Molecular/métodos , DNA Recombinante/síntese química , RNA Polimerases Dirigidas por DNA , Vírus da Encefalomiocardite/metabolismo , Elementos Facilitadores Genéticos , Genes Reporter , Vetores Genéticos/síntese química , Vetores Genéticos/metabolismo , Proteínas Luminescentes/síntese química , Proteínas Luminescentes/genética , Microscopia de Fluorescência por Excitação Multifotônica , Iniciação Traducional da Cadeia Peptídica/genética , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/genética , Retroviridae/genética , Transfecção , Proteínas Virais
18.
Methods Enzymol ; 429: 53-82, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17913619

RESUMO

A Krebs-2 cell-free extract that efficiently translates encephalomyocarditis virus (EMCV) RNA and extensively processes the viral polyprotein is also capable of supporting complete infectious EMCV replication. The system displays high RNA synthesis activity and de novo synthesis of virus up to titers of 2 x 10(7) to 6 x 10(7) plaque-forming units (pfu)/ml. The preparation of Krebs-2 cell extract and methods of analysis of EMCV-specific processes in vitro are described. We also demonstrate that the Krebs-2 cell-free system translates the entire open reading frame of the hepatitis C virus (HCV) RNA and properly processes the viral polyprotein when supplemented with canine microsomal membranes. In addition to processing, other posttranslational modifications of HCV proteins take place in vitro, such as the N-terminal glycosylation of the E1 and the E2 precursor (E2-p7) and phosphorylation of NS5A. The HCV RNA-programmed Krebs-2 cell-free extract should prove very useful as a novel screen for drugs that inhibit NS3-mediated processing. The use of this system should help fill the gap in understanding the regulation of synthesis and maturation of HCV proteins. With further optimization of cell-free conditions, the entire reconstitution of infectious HCV synthesis in vitro might become feasible.


Assuntos
Vírus da Encefalomiocardite/metabolismo , Genoma Viral/fisiologia , Hepacivirus/metabolismo , Biossíntese de Proteínas/fisiologia , RNA Viral/fisiologia , Replicação Viral/fisiologia , Animais , Carcinoma Krebs 2/metabolismo , Sistema Livre de Células , Feminino , Glicosilação , Camundongos , Proteínas do Envelope Viral/metabolismo
19.
J Virol Methods ; 142(1-2): 182-8, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17320977

RESUMO

Cell-free synthesis of an infectious virus is an ideal tool for elucidating the mechanism of viral replication and for screening anti-viral drugs. In the present study, the synthesis of Encephalomyocarditis virus (EMCV) from its RNA in HeLa and 293-F cell extracts was enhanced by employing a dialysis system in combination with a ribozyme technology. Although translation and processing of the EMCV polyprotein were not accelerated greatly by the dialysis system, de novo synthesis of viral RNA was enhanced considerably by dialysis, leading to a greater than eight-fold increased titer of synthesized EMCV compared with a conventional batch system. Furthermore, a synthetic EMCV RNA with a hammerhead ribozyme sequence at its 5'-end served as an efficient template for viral synthesis in the dialysis system. Therefore, this system provides opportunities for mutational analyses of EMCV in vitro.


Assuntos
Vírus da Encefalomiocardite/metabolismo , RNA Viral/metabolismo , Virologia/métodos , Linhagem Celular , Sistema Livre de Células , Diálise , Vírus da Encefalomiocardite/genética , Células HeLa , Humanos , Mutação , Biossíntese de Proteínas , RNA Catalítico , RNA Viral/síntese química
20.
Science ; 314(5801): 997-1001, 2006 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-17038589

RESUMO

Double-stranded RNA (dsRNA) produced during viral replication is believed to be the critical trigger for activation of antiviral immunity mediated by the RNA helicase enzymes retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5). We showed that influenza A virus infection does not generate dsRNA and that RIG-I is activated by viral genomic single-stranded RNA (ssRNA) bearing 5'-phosphates. This is blocked by the influenza protein nonstructured protein 1 (NS1), which is found in a complex with RIG-I in infected cells. These results identify RIG-I as a ssRNA sensor and potential target of viral immune evasion and suggest that its ability to sense 5'-phosphorylated RNA evolved in the innate immune system as a means of discriminating between self and nonself.


Assuntos
RNA Helicases DEAD-box/metabolismo , Imunidade Inata , Vírus da Influenza A/genética , Vírus da Influenza A/imunologia , RNA Viral/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Citoplasma/metabolismo , Citoplasma/virologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Células Dendríticas/virologia , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/imunologia , Vírus da Encefalomiocardite/metabolismo , Genoma Viral , Humanos , Vírus da Influenza A/metabolismo , Vírus da Influenza A/fisiologia , Interferon-alfa/biossíntese , Interferon beta/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Fosfatos/metabolismo , Fosforilação , Capuzes de RNA/metabolismo , RNA de Cadeia Dupla/metabolismo , RNA Viral/química , RNA Viral/genética , Receptores Imunológicos , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA