Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
1.
J Virol ; 96(17): e0112122, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-36000840

RESUMO

Seneca Valley virus (SVV) is a new pathogen associated with porcine idiopathic vesicular disease (PIVD) in recent years. However, SVV-host interaction is still unclear. In this study, through LC-MS/MS analysis and coimmunoprecipitation analysis, DHX30 was identified as a 3Cpro-interacting protein. 3Cpro mediated the cleavage of DHX30 at a specific site, which depends on its protease activity. Further study showed that DHX30 was an intrinsic antiviral factor against SVV that was dependent on its helicase activity. DHX30 functioned as a viral-RNA binding protein that inhibited SVV replication at the early stage of viral infection. RIP-seq showed comparatively higher coverage depth at SVV 5'UTR, but the distribution across SVV RNA suggested that the interaction had low specificity. DHX30 expression strongly inhibited double-stranded RNA (dsRNA) production. Interestingly, DHX30 was determined to interact with 3D in an SVV RNA-dependent manner. Thus, DHX30 negatively regulated SVV propagation by blocking viral RNA synthesis, presumably by participating in the viral replication complex. IMPORTANCE DHX30, an RNA helicase, is identified as a 3Cpro-interacting protein regulating Seneca Valley virus (SVV) replication dependent on its helicase activity. DHX30 functioned as a viral-RNA binding protein that inhibited SVV replication at the early stage of virus infection. DHX30 expression strongly inhibited double-stranded RNA (dsRNA) production. In addition, 3Cpro abolished DHX30 antiviral effects by inducing DHX30 cleavage. Thus, DHX30 is an intrinsic antiviral factor that inhibits SVV replication.


Assuntos
Proteases Virais 3C , Picornaviridae , Proteólise , RNA Helicases , Proteases Virais 3C/metabolismo , Animais , Cromatografia Líquida , Imunoprecipitação , Picornaviridae/enzimologia , Picornaviridae/genética , Picornaviridae/crescimento & desenvolvimento , Picornaviridae/fisiologia , Ligação Proteica , RNA Helicases/antagonistas & inibidores , RNA Helicases/metabolismo , RNA de Cadeia Dupla/biossíntese , RNA Viral/biossíntese , Suínos/virologia , Doença Vesicular Suína/virologia , Espectrometria de Massas em Tandem , Replicação Viral
2.
Viruses ; 13(11)2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34835106

RESUMO

Senecavirus A (SVA) is a member of the genus Senecavirus of the family Picornaviridae. SVA-associated vesicular disease (SAVD) outbreaks have been extensively reported since 2014-2015. Characteristic symptoms include vesicular lesions on the snout and feet as well as lameness in adult pigs and even death in piglets. The capsid protein VP3, a structural protein of SVA, is involved in viral replication and genome packaging. Here, we developed and characterized a mouse monoclonal antibody (mAb) 3E9 against VP3. A motif 192GWFSLHKLTK201 was identified as the linear B-cell epitope recognized by mAb 3E9 by using a panel of GFP-tagged epitope polypeptides. Sequence alignments show that 192GWFSLHKLTK201 was highly conserved in all SVA strains. Subsequently, alanine (A)-scanning mutagenesis indicated that W193, F194, L196, and H197 were the critical residues recognized by mAb 3E9. Further investigation with indirect immunofluorescence assay indicated that the VP3 protein was present in the cytoplasm during SVA replication. In addition, the mAb 3E9 specifically immunoprecipitated the VP3 protein from SVA-infected cells. Taken together, our results indicate that mAb 3E9 could be a powerful tool to work on the function of the VP3 protein during virus infection.


Assuntos
Proteínas do Capsídeo/imunologia , Infecções por Picornaviridae/virologia , Picornaviridae , Doenças dos Suínos/virologia , Animais , Proteínas do Capsídeo/genética , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Picornaviridae/crescimento & desenvolvimento , Picornaviridae/imunologia , Alinhamento de Sequência , Suínos , Replicação Viral
3.
Virology ; 561: 98-106, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34182259

RESUMO

Despite the ongoing interest in virus discovery, little is known about the factors that shape communities of viruses within individual hosts. Here, we address how virus communities might be impacted by the age of the hosts they infect, using total RNA sequencing to reveal the RNA viromes of different age groups of Ruddy Turnstones (Arenaria interpres). From oropharyngeal and cloacal swabs we identified 14 viruses likely infecting birds, 11 of which were novel, including members of the Reoviridae, Astroviridae, and Picornaviridae. Strikingly, 12 viruses identified were from juvenile birds sampled in the first year of their life, compared to only two viruses in adult birds. Both viral abundance and alpha diversity were marginally higher in juvenile than adult birds. As well as informing studies of virus ecology, that host age might be associated with viral composition is an important consideration for the future surveillance of novel and emerging viruses.


Assuntos
Charadriiformes/virologia , Vírus de RNA , Viroma , Envelhecimento , Animais , Astroviridae/classificação , Astroviridae/genética , Astroviridae/crescimento & desenvolvimento , Charadriiformes/fisiologia , Cloaca/virologia , Vírus de RNA de Cadeia Dupla/classificação , Vírus de RNA de Cadeia Dupla/genética , Vírus de RNA de Cadeia Dupla/crescimento & desenvolvimento , Genoma Viral , Orofaringe/virologia , Filogenia , Picornaviridae/classificação , Picornaviridae/genética , Picornaviridae/crescimento & desenvolvimento , Vírus de RNA/classificação , Vírus de RNA/genética , Vírus de RNA/crescimento & desenvolvimento , Reoviridae/classificação , Reoviridae/genética , Reoviridae/crescimento & desenvolvimento , Transcriptoma
4.
Virus Res ; 292: 198232, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33207264

RESUMO

Senecavirus A (SVA), previously known as Seneca Valley virus, is classified into the genus Senecavirus in the family Picornaviridae. SVA is not pathogenic to normal human cells, but has potent oncolytic activity in some tumor cells with neuroendocrine feature, such as small cell lung cancer (SCLC) NCI-H446 cell line. In this study, we rescued and characterized a recombinant SVA that could efficiently express a novel luciferase, NanoLuc® luciferase (NLuc), which was smaller and "brighter" than others. This NLuc-tagged recombinant SVA (rSVA-NLuc) exhibited high capacity for viral replication, but genetic instability of NLuc during serial virus passages. The NLuc as a reporter facilitated oncolytic analysis of rSVA-NLuc in H446 cells. The rSVA-NLuc-infected H446 cells exhibited an oncolytic phenotype characterized by cell rounding, swelling, detachment and lysis at 48 h post infection. Kinetic curve showed that the NLuc was rapidly expressed in H446 cells during an exponential phase of viral growth. Because the NLuc offered several advantages over fluorescent proteins for assay scalability in vivo, the rSVA-NLuc would play a potential role in facilitating in vivo imaging studies of oncolytic virotherapy.


Assuntos
Luciferases/genética , Vírus Oncolíticos/genética , Picornaviridae/genética , Linhagem Celular Tumoral , Expressão Gênica , Humanos , Luciferases/metabolismo , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/crescimento & desenvolvimento , Vírus Oncolíticos/fisiologia , Picornaviridae/crescimento & desenvolvimento , Picornaviridae/fisiologia , Replicação Viral
5.
Front Immunol ; 11: 577838, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33133097

RESUMO

Stress granules (SGs) are the sites of mRNA storage and related to the regulation of mRNA translation, which are dynamic structures in response to various environmental stresses and viral infections. Seneca Valley virus (SVV), an oncolytic RNA virus belonging to Picornaviridae family, can cause vesicular disease (VD) indistinguished from foot-and-mouth disease (FMD) and other pig VDs. In this study, we found that SVV induced SG formation in the early stage of infection in a PKR-eIF2α dependent manner, as demonstrated by the recruitment of marker proteins of G3BP1 and eIF4GI. Surprisingly, we found that downregulating SG marker proteins TIA1 or G3BP1, or expressing an eIF2α non-phosphorylatable mutant inhibited SG formation, but this inhibition of transient SG formation had no significant effect on SVV propagation. Depletion of G3BP1 significantly attenuated the activation of NF-κB signaling pathway. In addition, we found that SVV inhibited SG formation at the late stage of infection and 3C protease was essential for the inhibition depending on its enzyme activity. Furthermore, we also found that 3C protease blocked the SG formation by disrupting eIF4GI-G3BP1 interaction. Overall, our results demonstrate that SVV induces transient SG formation in an eIF2α phosphorylation and PKR-dependent manner, and that 3C protease inhibits SG formation by interfering eIF4GI-G3BP1 interaction.


Assuntos
Proteases Virais 3C/metabolismo , Grânulos Citoplasmáticos/metabolismo , DNA Helicases/metabolismo , Fator de Iniciação Eucariótico 4G/metabolismo , Picornaviridae/enzimologia , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , RNA Helicases/metabolismo , Proteínas com Motivo de Reconhecimento de RNA/metabolismo , Estresse Fisiológico , Proteases Virais 3C/genética , Grânulos Citoplasmáticos/virologia , DNA Helicases/genética , Fator de Iniciação Eucariótico 4G/genética , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Fosforilação , Picornaviridae/genética , Picornaviridae/crescimento & desenvolvimento , Proteínas de Ligação a Poli-ADP-Ribose/genética , Ligação Proteica , RNA Helicases/genética , Proteínas com Motivo de Reconhecimento de RNA/genética , Transdução de Sinais , Replicação Viral , eIF-2 Quinase/metabolismo
6.
PLoS Pathog ; 16(10): e1008970, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33104725

RESUMO

Senecavirus A (SVA), discovered in 2002, is an emerging pathogen of swine that has since been reported in numerous pork producing countries. To date, the mechanism of SVA replication remains poorly understood. In this study, utilizing iTRAQ analysis we found that UBE2L6, an E2 ubiquitin-conjugating enzyme, is up-regulated in SVA-infected BHK-21 cells, and that its overexpression promotes SVA replication. We determined that UBE2L6 interacts with, and ubiquitinates the RNA-dependent RNA polymerase of SVA, (the 3D protein) and this ubiquitination serves to inhibit the degradation of 3D. UBE2L6-mediated ubiquitination of 3D requires a cystine at residue 86 in UBE2L6, and lysines at residues 169 and 321 in 3D. Virus with mutations in 3D (rK169R and rK321R) exhibited significantly decreased replication compared to wild type SVA and the repaired viruses, rK169R(R) and rK321R(R). These data indicate that UBE2L6, the enzyme, targets the 3D polymerase, the substrate, during SVA infection to facilitate replication.


Assuntos
Infecções por Picornaviridae/virologia , Picornaviridae/crescimento & desenvolvimento , RNA Polimerase Dependente de RNA/química , Enzimas de Conjugação de Ubiquitina/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Animais , Células Cultivadas , Cricetinae , Interações Hospedeiro-Patógeno , Picornaviridae/enzimologia , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/metabolismo , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Suínos , Enzimas de Conjugação de Ubiquitina/genética , Ubiquitinação , Proteínas Virais/genética
7.
J Cell Mol Med ; 24(12): 6988-6999, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32374474

RESUMO

Outbreaks of infections with viruses like Sars-CoV-2, Ebola virus and Zika virus lead to major global health and economic problems because of limited treatment options. Therefore, new antiviral drug candidates are urgently needed. The promising new antiviral drug candidate silvestrol effectively inhibited replication of Corona-, Ebola-, Zika-, Picorna-, Hepatis E and Chikungunya viruses. Besides a direct impact on pathogens, modulation of the host immune system provides an additional facet to antiviral drug development because suitable immune modulation can boost innate defence mechanisms against the pathogens. In the present study, silvestrol down-regulated several pro- and anti-inflammatory cytokines (IL-6, IL-8, IL-10, CCL2, CCL18) and increased TNF-α during differentiation and activation of M1-macrophages, suggesting that the effects of silvestrol might cancel each other out. However, silvestrol amplified the anti-inflammatory potential of M2-macrophages by increasing expression of anti-inflammatory surface markers CD206, TREM2 and reducing release of pro-inflammatory IL-8 and CCL2. The differentiation of dendritic cells in the presence of silvestrol is characterized by down-regulation of several surface markers and cytokines indicating that differentiation is impaired by silvestrol. In conclusion, silvestrol influences the inflammatory status of immune cells depending on the cell type and activation status.


Assuntos
Antivirais/farmacologia , Betacoronavirus/efeitos dos fármacos , Citocinas/genética , Células Dendríticas/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Macrófagos/efeitos dos fármacos , Triterpenos/farmacologia , Betacoronavirus/crescimento & desenvolvimento , Betacoronavirus/imunologia , Diferenciação Celular/efeitos dos fármacos , Vírus Chikungunya/efeitos dos fármacos , Vírus Chikungunya/crescimento & desenvolvimento , Vírus Chikungunya/imunologia , Citocinas/classificação , Citocinas/imunologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Ebolavirus/efeitos dos fármacos , Ebolavirus/crescimento & desenvolvimento , Ebolavirus/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Vírus da Hepatite E/efeitos dos fármacos , Vírus da Hepatite E/crescimento & desenvolvimento , Vírus da Hepatite E/imunologia , Humanos , Imunidade Inata/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/virologia , Especificidade de Órgãos , Picornaviridae/efeitos dos fármacos , Picornaviridae/crescimento & desenvolvimento , Picornaviridae/imunologia , Cultura Primária de Células , SARS-CoV-2 , Transdução de Sinais , Zika virus/efeitos dos fármacos , Zika virus/crescimento & desenvolvimento , Zika virus/imunologia
8.
Viruses ; 12(2)2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-32023921

RESUMO

In this study, we characterized the role of host cell protein tyrosyl-DNA phosphodiesterase 2 (TDP2) activity, also known as VPg unlinkase, in picornavirus infections in a human cell model of infection. TDP2/VPg unlinkase is used by picornaviruses to remove the small polypeptide, VPg (Virus Protein genome-linked, the primer for viral RNA synthesis), from virus genomic RNA. We utilized a CRISPR/Cas-9-generated TDP2 knock out (KO) human retinal pigment epithelial-1 (hRPE-1) cell line, in addition to the wild type (WT) counterpart for our studies. We determined that in the absence of TDP2, virus growth kinetics for two enteroviruses (poliovirus and coxsackievirus B3) were delayed by about 2 h. Virus titers were reduced by ~2 log10 units for poliovirus and 0.5 log10 units for coxsackievirus at 4 hours post-infection (hpi), and by ~1 log10 unit at 6 hpi for poliovirus. However, virus titers were nearly indistinguishable from those of control cells by the end of the infectious cycle. We determined that this was not the result of an alternative source of VPg unlinkase activity being activated in the absence of TPD2 at late times of infection. Viral protein production in TDP2 KO cells was also substantially reduced at 4 hpi for poliovirus infection, consistent with the observed growth kinetics delay, but reached normal levels by 6 hpi. Interestingly, this result differs somewhat from what has been reported previously for the TDP2 KO mouse cell model, suggesting that either cell type or species-specific differences might be playing a role in the observed phenotype. We also determined that catalytically inactive TDP2 does not rescue the growth defect, confirming that TDP2 5' phosphodiesterase activity is required for efficient virus replication. Importantly, we show for the first time that polysomes can assemble efficiently on VPg-linked RNA after the initial round of translation in a cell culture model, but both positive and negative strand RNA production is impaired in the absence of TDP2 at mid-times of infection, indicating that the presence of VPg on the viral RNA affects a step in the replication cycle downstream of translation (e.g., RNA synthesis). In agreement with this conclusion, we found that double-stranded RNA production (a marker of viral RNA synthesis) is delayed in TDP2 KO RPE-1 cells. Moreover, we show that premature encapsidation of nascent, VPg-linked RNA is not responsible for the observed virus growth defect. Our studies provide the first lines of evidence to suggest that either negative- or positive-strand RNA synthesis (or both) is a likely candidate for the step that requires the removal of VPg from the RNA for an enterovirus infection to proceed efficiently.


Assuntos
Proteínas de Ligação a DNA/genética , Diester Fosfórico Hidrolases/genética , Infecções por Picornaviridae/virologia , Picornaviridae/genética , Sistemas CRISPR-Cas , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/virologia , Técnicas de Inativação de Genes , Humanos , Diester Fosfórico Hidrolases/metabolismo , Picornaviridae/crescimento & desenvolvimento , Polirribossomos/imunologia , RNA de Cadeia Dupla/genética , RNA Viral/genética , Retina/citologia
9.
Elife ; 82019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31769754

RESUMO

Comprehensive knowledge of the host factors required for picornavirus infection would facilitate antiviral development. Here we demonstrate roles for three human genes, TNK2, WASL, and NCK1, in infection by multiple picornaviruses. CRISPR deletion of TNK2, WASL, or NCK1 reduced encephalomyocarditis virus (EMCV), coxsackievirus B3 (CVB3), poliovirus and enterovirus D68 infection, and chemical inhibitors of TNK2 and WASL decreased EMCV infection. Reduced EMCV lethality was observed in mice lacking TNK2. TNK2, WASL, and NCK1 were important in early stages of the viral lifecycle, and genetic epistasis analysis demonstrated that the three genes function in a common pathway. Mechanistically, reduced internalization of EMCV was observed in TNK2 deficient cells demonstrating that TNK2 functions in EMCV entry. Domain analysis of WASL demonstrated that its actin nucleation activity was necessary to facilitate viral infection. Together, these data support a model wherein TNK2, WASL, and NCK1 comprise a pathway important for multiple picornaviruses.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Interações Hospedeiro-Patógeno , Proteínas Oncogênicas/metabolismo , Picornaviridae/crescimento & desenvolvimento , Proteínas Tirosina Quinases/metabolismo , Internalização do Vírus , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Animais , Infecções por Cardiovirus/patologia , Linhagem Celular , Modelos Animais de Doenças , Deleção de Genes , Humanos , Camundongos Knockout , Proteínas Oncogênicas/deficiência , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/deficiência , Análise de Sobrevida , Proteína Neuronal da Síndrome de Wiskott-Aldrich/antagonistas & inibidores , Proteína Neuronal da Síndrome de Wiskott-Aldrich/deficiência
10.
Virology ; 537: 254-263, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31539773

RESUMO

Diverse effects on autophagy, a cell degradation pathway, have been associated with the infectious mechanisms of different pathogens. Here, we demonstrated that Seneca valley virus (SVV), an important emerging porcine virus characterized by vesicular lesions and neonatal mortality, can induce autophagy in cultured PK-15 and BHK-21 cells by detecting autophagosome formation, GFP-LC3 puncta and accumulation of LC3-II proteins. Treatment with pharmacological inducers/inhibitors and small interfering RNA sequences targeting genes critical for autophagosome formation affected autophagy induction and viral yields. SVV induced a complete autophagic process to enhance its replication. The PERK and ATF6 pathways, two components of the endoplasmic reticulum (ER)-related unfolded protein response (UPR), were also activated in SVV-infected cells and downregulation of their expression suppressed SVV-induced autophagy and viral yields. Overall, these results reveal that SVV induces autophagy in cultured cells through the PERK and ATF6 pathways, thereby contributing to understanding of the molecular mechanisms underlying SVV pathogenesis.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Autofagia , Interações Hospedeiro-Patógeno , Picornaviridae/crescimento & desenvolvimento , Resposta a Proteínas não Dobradas , eIF-2 Quinase/metabolismo , Animais , Autofagossomos/metabolismo , Linhagem Celular , Cricetinae , Suínos , Replicação Viral
11.
Virology ; 535: 122-129, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31299488

RESUMO

Seneca Valley virus (SVV) is a member of the Picornaviridae family, which has been used to treat neuroendocrine cancer. The innate immune system plays an important role in SVV infection. However, few studies have elucidated the relationship between SVV infection and the host's antiviral response. In this study, SVV replication could induce the degradation of RIG-I in HEK-293T, SW620 and SK6 cells. And overexpressing retinoic acid-inducible gene I (RIG-I) could significantly inhibit SVV propagation. The viral protein 2C and 3C were essential for the degradation of RIG-I. Furthermore, 2C and 3C significantly reduced Sev or RIG-I-induced IFN-ß production. Mechanistically, 2C and 3C induced RIG-I degradation through the caspase signaling pathway. Taken together, we demonstrate the antiviral role of RIG-I against SVV and the mechanism by which SVV 2C and 3C weaken the host innate immune system.


Assuntos
Proteína DEAD-box 58/metabolismo , Interações entre Hospedeiro e Microrganismos , Evasão da Resposta Imune , Interferon Tipo I/antagonistas & inibidores , Picornaviridae/imunologia , Proteólise , Proteínas Virais/metabolismo , Linhagem Celular , Humanos , Picornaviridae/crescimento & desenvolvimento , Receptores Imunológicos , Transdução de Sinais , Replicação Viral
12.
Curr Opin Virol ; 37: 58-62, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31284078

RESUMO

Enteric viruses infect the mammalian gastrointestinal tract which is home to a diverse community of intestinal bacteria. Accumulating evidence suggests that certain enteric viruses utilize these bacteria to promote infection. While this is not surprising considering their proximity, multiple viruses from different viral families have been shown to bind directly to bacteria or bacterial components to aid in viral replication, pathogenesis, and transmission. These data suggest that the concept of a single virus infecting a single cell, independent of the environment, needs to be reevaluated. In this review, I will discuss the current knowledge of enteric virus-bacterial interactions and discuss the implications for viral pathogenesis and transmission.


Assuntos
Trato Gastrointestinal/virologia , Interações Microbianas , Microbiota , Vírus , Animais , Trato Gastrointestinal/microbiologia , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Evasão da Resposta Imune , Picornaviridae/crescimento & desenvolvimento , Picornaviridae/patogenicidade , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/microbiologia , Infecções por Picornaviridae/transmissão , Reoviridae/crescimento & desenvolvimento , Reoviridae/patogenicidade , Infecções por Reoviridae/imunologia , Infecções por Reoviridae/microbiologia , Infecções por Reoviridae/transmissão , Retroviridae/crescimento & desenvolvimento , Retroviridae/patogenicidade , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/microbiologia , Infecções por Retroviridae/transmissão , Viroses/imunologia , Viroses/microbiologia , Viroses/transmissão , Replicação Viral , Vírus/crescimento & desenvolvimento , Vírus/patogenicidade
13.
Food Environ Virol ; 11(2): 193-197, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30895522

RESUMO

Salivirus is a newly discovered virus which seems to be related to acute gastroenteritis in children. Salivirus may infect susceptible children by fecal-oral route after exposure to contaminated water. The present study aims to evaluate the occurrence and quantity of Salivirus in treated and untreated sewage water and river water samples collected in the city of Karaj, Iran by reverse transcription-quantitative PCR assay. A total of 50 samples were collected from environmental waters containing 22 treated and untreated sewage water in volume of 1 l and 28 river water samples in volume of 5 l were included in this study. After viral RNA extraction, the Real-time PCR was performed to amplify the 5'UTR sequence of Salivirus genome and viral load was assessed. Out of the 50 samples tested, the Salivirus genomic RNA was identified in 5/12 (41.6%) of treated and 3/10 (30%) of untreated sewage samples and in 8/28 (28.5%) of river water samples. The maximum viral load was 4.8 × 106 copies/l in treated sewage water sample in September and the lower viral load was 4 × 105 copies/l related to treated sewage water taken in December. This is the first report of Salivirus occurrence in the environmental waters in Iran. The viral prevalence of Salivirus in each of the three sets of tested samples was within low to moderate in range.


Assuntos
Picornaviridae/isolamento & purificação , Rios/virologia , Esgotos/virologia , Genoma Viral , Irã (Geográfico) , Picornaviridae/classificação , Picornaviridae/genética , Picornaviridae/crescimento & desenvolvimento , Poluição da Água/análise
14.
Antiviral Res ; 160: 183-189, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30408499

RESUMO

The mechanisms that enable Seneca Valley Virus (SVV) to escape the host innate immune response are not well known. Previous studies demonstrated that SVV 3Cpro suppresses innate immune responses by cleavage of host proteins and degradation of IRF3 and IRF7 protein expression. Here, we showed that SVV 3C protease (3Cpro) has deubiquitinating activity. Overexpressed 3Cpro inhibits the ubiquitination of cellular substrates, acting on both lysine-48- and lysine-63-linked polyubiquitin chains. SVV infection also possessed deubiquitinating activity. The ubiquitin-proteasome system was significantly involved in SVV replication. Furthermore, 3Cpro inhibited the ubiquitination of retinoic acid-inducible gene I (RIG-I), TANK-binding kinase 1 (TBK1), and TNF receptor-associated factor 3 (TRAF3), thereby blocking the expression of interferon (IFN)-ß and IFN stimulated gene 54 (ISG54) mRNAs. A detailed analysis revealed that mutations (H48A, C160A, or H48A/C160A) that ablate the Cys and His residues of 3Cpro abrogated its deubiquitinating activity and the ability of 3Cpro to block IFN-ß induction. Together, our results demonstrate a novel mechanism developed by SVV 3Cpro to promote viral replication, and may also provide a novel strategy for improving ubiquitination-based therapy.


Assuntos
Cisteína Endopeptidases/metabolismo , Enzimas Desubiquitinantes/metabolismo , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Picornaviridae/imunologia , Picornaviridae/patogenicidade , Proteínas Virais/metabolismo , Proteases Virais 3C , Substituição de Aminoácidos , Análise Mutacional de DNA , Fatores Imunológicos/metabolismo , Picornaviridae/crescimento & desenvolvimento , Replicação Viral
15.
Viruses ; 9(7)2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28671611

RESUMO

Senecavirus A (SVA) is a positive-sense single-stranded RNA virus that belongs to the Senecavirus genus within the Picornaviridae family. The virus has been silently circulating in pig herds of the USA since 1988. However, cases of senecavirus-associated vesicular disease were reported in Canada in 2007 and in the USA in 2012. Since late 2014 and early 2015, an increasing number of senecavirus outbreaks have been reported in pigs in different producing categories, with this virus being detected in Brazil, China, and Thailand. Considering the novel available data on senecavirus infection and disease, 2015 may be a divisor in the epidemiology of the virus. Among the aspects that reinforce this hypothesis are the geographical distribution of the virus, the affected pig-producing categories, clinical signs associated with the infection, and disease severity. This review presents the current knowledge regarding the senecavirus infection and disease, especially in the last two years. Senecavirus epidemiology, pathogenic potential, host immunological response, diagnosis, and prophylaxis and control measures are addressed. Perspectives are focused on the need for complete evolutionary, epidemiological and pathogenic data and the capability for an immediate diagnosis of senecavirus infection. The health risks inherent in the swine industry cannot be neglected.


Assuntos
Infecções por Picornaviridae/veterinária , Picornaviridae/crescimento & desenvolvimento , Doenças dos Suínos/epidemiologia , Doenças dos Suínos/virologia , América/epidemiologia , Animais , Ásia/epidemiologia , Controle de Doenças Transmissíveis/métodos , Testes Diagnósticos de Rotina , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Suínos , Doenças dos Suínos/patologia
16.
PLoS One ; 12(7): e0180910, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28686725

RESUMO

Deformed wing virus (DWV) is a lethal virus of honeybees (Apis mellifera) implicated in elevated colony mortality rates worldwide and facilitated through vector transmission by the ectoparasitic mite Varroa destructor. Clinical, symptomatic DWV infections are almost exclusively associated with high virus titres during pupal development, usually acquired through feeding by Varroa mites when reproducing on bee pupae. Control of the mite population, generally through acaricide treatment, is essential for breaking the DWV epidemic and minimizing colony losses. In this study, we evaluated the effectiveness of remedial mite control on clearing DWV from a colony. DWV titres in adult bees and pupae were monitored at 2 week intervals through summer and autumn in acaricide-treated and untreated colonies. The DWV titres in Apistan treated colonies was reduced 1000-fold relative to untreated colonies, which coincided with both the removal of mites and also a turnover of the bee population in the colony. This adult bee population turnover is probably more critical than previously realized for effective clearing of DWV infections. After this initial reduction, subclinical DWV titres persisted and even increased again gradually during autumn, demonstrating that alternative non-Varroa transmission routes can maintain the DWV titres at significant subclinical levels even after mite removal. The implications of these results for practical recommendations to mitigate deleterious subclinical DWV infections and improving honeybee health management are discussed.


Assuntos
Acaricidas/farmacologia , Abelhas/virologia , Ectoparasitoses/prevenção & controle , Picornaviridae/efeitos dos fármacos , Controle de Ácaros e Carrapatos/métodos , Varroidae/efeitos dos fármacos , Animais , Abelhas/parasitologia , Vetores de Doenças , Ectoparasitoses/parasitologia , Ectoparasitoses/virologia , Picornaviridae/crescimento & desenvolvimento , Picornaviridae/patogenicidade , Pupa/parasitologia , Pupa/virologia , RNA Viral/genética , Estações do Ano , Varroidae/virologia , Carga Viral/efeitos dos fármacos
17.
J Virol ; 91(16)2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28566380

RESUMO

Seneca Valley virus (SVV) is an oncolytic RNA virus belonging to the Picornaviridae family. Its nucleotide sequence is highly similar to those of members of the Cardiovirus genus. SVV is also a neuroendocrine cancer-selective oncolytic picornavirus that can be used for anticancer therapy. However, the interaction between SVV and its host is yet to be fully characterized. In this study, SVV inhibited antiviral type I interferon (IFN) responses by targeting different host adaptors, including mitochondrial antiviral signaling (MAVS), Toll/interleukin 1 (IL-1) receptor domain-containing adaptor inducing IFN-ß (TRIF), and TRAF family member-associated NF-κB activator (TANK), via viral 3C protease (3Cpro). SVV 3Cpro mediated the cleavage of MAVS, TRIF, and TANK at specific sites, which required its protease activity. The cleaved MAVS, TRIF, and TANK lost the ability to regulate pattern recognition receptor (PRR)-mediated IFN production. The cleavage of TANK also facilitated TRAF6-induced NF-κB activation. SVV was also found to be sensitive to IFN-ß. Therefore, SVV suppressed antiviral IFN production to escape host antiviral innate immune responses by cleaving host adaptor molecules.IMPORTANCE Host cells have developed various defenses against microbial pathogen infection. The production of IFN is the first line of defense against microbial infection. However, viruses have evolved many strategies to disrupt this host defense. SVV, a member of the Picornavirus genus, is an oncolytic virus that shows potential functions in anticancer therapy. It has been demonstrated that IFN can be used in anticancer therapy for certain tumors. However, the relationship between oncolytic virus and innate immune response in anticancer therapy is still not well known. In this study, we showed that SVV has evolved as an effective mechanism to inhibit host type I IFN production by using its 3Cpro to cleave the molecules MAVS, TRIF, and TANK directly. These molecules are crucial for the Toll-like receptor 3 (TLR3)-mediated and retinoic acid-inducible gene I (RIG-I)-like receptor (RLR)-mediated signaling pathway. We also found that SVV is sensitive to IFN-ß. These findings increase our understanding of the interaction between SVV and host innate immunity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Cisteína Endopeptidases/metabolismo , Evasão da Resposta Imune , Interferon Tipo I/antagonistas & inibidores , Picornaviridae/crescimento & desenvolvimento , Proteínas Virais/metabolismo , Proteases Virais 3C , Animais , Linhagem Celular , Cricetinae , Interações Hospedeiro-Patógeno , Humanos , Picornaviridae/enzimologia , Proteólise
18.
J Vis Exp ; (120)2017 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-28287517

RESUMO

Cell-specific restriction of viral replication without concomitant attenuation can benefit vaccine development, gene therapy, oncolytic virotherapy, and understanding the biological properties of viruses. There are several mechanisms for regulating viral tropism, however they tend to be virus class specific and many result in virus attenuation. Additionally, many viruses, including picornaviruses, exhibit size constraints that do not allow for incorporation of large amounts of foreign genetic material required for some targeting methods. MicroRNAs are short, non-coding RNAs that regulate gene expression in eukaryotic cells by binding complementary target sequences in messenger RNAs, preventing their translation or accelerating their degradation. Different cells exhibit distinct microRNA signatures and many microRNAs serve as biomarkers. These differential expression patterns can be exploited for restricting gene expression in cells that express specific microRNAs while maintaining expression in cells that do not. In regards to regulating viral tropism, sequences complementary to specific microRNAs are incorporated into the viral genome, generally in the 3' non-coding regions, targeting them for destruction in the presence of the cognate microRNAs thus preventing viral gene expression and/or replication. MicroRNA-targeting is a technique that theoretically can be applied to all viral vectors without altering the potency of the virus in the absence of the corresponding microRNAs. Here we describe experimental methods associated with generating a microRNA-targeted picornavirus and evaluating the efficacy and specificity of that targeting in vitro. This protocol is designed for a rapidly replicating virus with a lytic replication cycle, however, modification of the time points analyzed and the specific virus titration readouts used will aid in the adaptation of this protocol to many different viruses.


Assuntos
Genoma Viral , MicroRNAs/genética , Terapia Viral Oncolítica/métodos , Infecções por Picornaviridae/virologia , Picornaviridae/genética , RNA Viral/genética , Replicação Viral/genética , Vetores Genéticos , Humanos , Picornaviridae/crescimento & desenvolvimento , Tropismo
19.
Curr Opin Virol ; 19: 1-6, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27240115

RESUMO

Picornavirus infection induces rapid reorganization of the cellular membrane architecture and appearance of novel membranous structures associated with the viral RNA replication and virion assembly-replication organelles. Recent studies significantly advanced our understanding of their lipid composition and cellular mechanisms involved in their development. Picornaviruses activate synthesis of both structural and signaling lipids and reroute cellular cholesterol trafficking pathways to create unique membranous domains favoring viral replication. Rapidly replicating picornaviruses rely on posttranslational activation and/or specific recruitment of cellular proteins rather than on modulation of expression of cellular genes to create favorable membrane microenvironment. At the same time picornaviruses demonstrate remarkable adaptability to changes in the lipid landscape which should be taken into account when developing novel antiviral strategies.


Assuntos
Organelas/química , Picornaviridae/fisiologia , Replicação Viral , Membrana Celular/química , Membrana Celular/fisiologia , Replicação do DNA , Humanos , Metabolismo dos Lipídeos , Lipídeos/química , Organelas/fisiologia , Organelas/ultraestrutura , Picornaviridae/genética , Picornaviridae/crescimento & desenvolvimento , RNA Viral
20.
Viral Immunol ; 29(6): 343-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27092424

RESUMO

Intestinal epithelial cells, which serve as the first physical barrier to protect intestinal tract from external antigens, have an important role in the local innate immunity. Screening of reference genes that have stable expression levels after viral infection in porcine intestinal epithelial cells is critical for ensuring the reliability of the expression analysis on anti-infection genes in porcine intestinal epithelial cells. In this study, nine common reference genes in pigs, including ACTB, B2M, GAPDH, HMBS, SDHA, HPRT1, TBP, YWHAZ, and RPL32, were chosen as the candidate reference genes. Porcine sapelovirus (PSV) was used as a model virus to infect porcine intestinal epithelial cell line (IPEC-J2). The expression stability of the nine genes was assessed by the geNorm, NormFinder, and BestKeeper software. Moreover, RefFinder program was used to evaluate the analytical results of above three softwares, and a relative expression experiment of selected target gene was used to verify the analysis results. The comprehensive results indicated that the gene combination of TBP and RPL32 has the most stable expression, which could be considered as an appropriate reference gene for research on gene expression after PSV infection in IPEC-J2cells. The results provided essential data for expression analysis of anti-infection genes in porcine intestinal epithelial cells.


Assuntos
Células Epiteliais/virologia , Perfilação da Expressão Gênica/normas , Interações Hospedeiro-Patógeno , Picornaviridae/crescimento & desenvolvimento , Padrões de Referência , Animais , Linhagem Celular , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA