Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 34(9): 108798, 2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33657363

RESUMEN

Type I interferons (IFNs) induce hundreds of IFN-stimulated genes (ISGs) in response to viral infection. Induction of these ISGs must be regulated for an efficient and controlled antiviral response, but post-transcriptional controls of these genes have not been well defined. Here, we identify a role for the RNA base modification N6-methyladenosine (m6A) in the regulation of ISGs. Using ribosome profiling and quantitative mass spectrometry, coupled with m6A-immunoprecipitation and sequencing, we identify a subset of ISGs, including IFITM1, whose translation is enhanced by m6A and the m6A methyltransferase proteins METTL3 and METTL14. We further determine that the m6A reader YTHDF1 increases the expression of IFITM1 in an m6A-binding-dependent manner. Importantly, we find that the m6A methyltransferase complex promotes the antiviral activity of type I IFN. Thus, these studies identify m6A as having a role in post-transcriptional control of ISG translation during the type I IFN response for antiviral restriction.


Asunto(s)
Adenosina/análogos & derivados , Biosíntesis de Proteínas , Procesamiento Postranscripcional del ARN , Transcripción Genética , Estomatitis Vesicular/genética , Vesiculovirus/patogenicidad , Células A549 , Adenosina/metabolismo , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Antivirales/farmacología , Chlorocebus aethiops , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Interferón beta/farmacología , Metiltransferasas/biosíntesis , Metiltransferasas/genética , Biosíntesis de Proteínas/efectos de los fármacos , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transcripción Genética/efectos de los fármacos , Células Vero , Estomatitis Vesicular/metabolismo , Estomatitis Vesicular/virología , Vesiculovirus/crecimiento & desarrollo , Replicación Viral
2.
Front Immunol ; 11: 607889, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33584680

RESUMEN

Early and strong production of IFN-I by dendritic cells is important to control vesicular stomatitis virus (VSV), however mechanisms which explain this cell-type specific innate immune activation remain to be defined. Here, using a genome wide association study (GWAS), we identified Integrin alpha-E (Itgae, CD103) as a new regulator of antiviral IFN-I production in a mouse model of vesicular stomatitis virus (VSV) infection. CD103 was specifically expressed by splenic conventional dendritic cells (cDCs) and limited IFN-I production in these cells during VSV infection. Mechanistically, CD103 suppressed AKT phosphorylation and mTOR activation in DCs. Deficiency in CD103 accelerated early IFN-I in cDCs and prevented death in VSV infected animals. In conclusion, CD103 participates in regulation of cDC specific IFN-I induction and thereby influences immune activation after VSV infection.


Asunto(s)
Antígenos CD/metabolismo , Células Dendríticas/virología , Inmunidad Innata , Cadenas alfa de Integrinas/metabolismo , Interferón Tipo I/metabolismo , Estomatitis Vesicular/virología , Vesiculovirus/patogenicidad , Animales , Antígenos CD/genética , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Estudio de Asociación del Genoma Completo , Interacciones Huésped-Patógeno , Cadenas alfa de Integrinas/genética , Ratones de la Cepa 129 , Ratones Endogámicos AKR , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos NOD , Ratones Noqueados , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Estomatitis Vesicular/genética , Estomatitis Vesicular/inmunología , Estomatitis Vesicular/metabolismo , Vesiculovirus/crecimiento & desarrollo , Replicación Viral
3.
Nat Cell Biol ; 21(11): 1346-1356, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31685995

RESUMEN

In the past two decades, emerging studies have suggested that DExD/H box helicases belonging to helicase superfamily 2 (SF2) play essential roles in antiviral innate immunity. However, the antiviral functions of helicase SF1, which shares a conserved helicase core with SF2, are little understood. Here we demonstrate that zinc finger NFX1-type containing 1 (ZNFX1), a helicase SF1, is an interferon (IFN)-stimulated, mitochondrial-localised dsRNA sensor that specifically restricts the replication of RNA viruses. Upon virus infection, ZNFX1 immediately recognizes viral RNA through its Armadillo-type fold and P-loop domain and then interacts with mitochondrial antiviral signalling protein to initiate the type I IFN response without depending on retinoic acid-inducible gene I-like receptors (RLRs). In short, as is the case with interferon-stimulated genes (ISGs) alone, ZNFX1 can induce IFN and ISG expression at an early stage of RNA virus infection to form a positively regulated loop of the well-known RLR signalling. This provides another layer of understanding of the complexity of antiviral immunity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Antígenos de Neoplasias/genética , Mitocondrias/inmunología , Factores de Empalme de ARN/genética , ARN Bicatenario/genética , ARN Viral/genética , Proteínas de Unión al ARN/metabolismo , Factores Generales de Transcripción/metabolismo , Vesiculovirus/genética , Células A549 , Proteínas Adaptadoras Transductoras de Señales/inmunología , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/inmunología , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/inmunología , Regulación de la Expresión Génica , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/virología , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/virología , Conformación de Ácido Nucleico , Poli I-C/farmacología , Cultivo Primario de Células , Unión Proteica , Factores de Empalme de ARN/inmunología , ARN Bicatenario/química , ARN Bicatenario/inmunología , ARN Viral/química , ARN Viral/inmunología , Proteínas de Unión al ARN/genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Factores Generales de Transcripción/genética , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/inmunología
4.
J Gen Virol ; 100(11): 1478-1490, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31553299

RESUMEN

Vesicular stomatitis (VS) is a notifiable disease of livestock affecting cattle, horses, pigs and humans. Vesicular stomatitis virus (VSV) serotypes Indiana and New Jersey are endemic to Central America; however, they also cause sporadic and scattered outbreaks in various countries in South and North America, including the USA. In order to develop an effective experimental challenge model for VSV, we compared the pathogenicity of three VSV serotype Indiana isolates in 36 4-5 week-old pigs. Two bovine isolates of Central American origin and one equine isolate from the USA were used for the experimental infections. Each pig was inoculated with a single isolate by both the intradermal and intranasal routes. Clinical signs of VSV infection were recorded daily for 10 days post-inoculation (days p.i.). Nasal and tonsillar swab samples and blood were collected to monitor immune responses, virus replication and shedding. Post-challenge, characteristic signs of VS were observed, including vesicles on the nasal planum and coronary bands, lameness, loss of hoof walls and pyrexia. Pigs inoculated with the Central American isolates showed consistently more severe clinical signs in comparison to the pigs infected with the USA isolate. Genomic RNA was isolated from the original challenge virus stocks, sequenced and compared to VSV genomes available in GenBank. Comparative genome analysis demonstrated significant differences between the VSV isolate from the USA and the two Central American isolates. Our results indicate that the Central American isolates of VSV serotype Indiana used in this study are more virulent in swine than the USA VSV serotype Indiana isolate and represent good candidate challenge strains for future VSV studies.


Asunto(s)
Modelos Animales de Enfermedad , Estomatitis Vesicular/patología , Estomatitis Vesicular/virología , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/patogenicidad , Estructuras Animales/patología , Estructuras Animales/virología , Animales , Sangre/virología , Serogrupo , Porcinos , Vesiculovirus/clasificación , Virulencia , Replicación Viral , Esparcimiento de Virus
5.
J Biosci ; 44(2)2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31180044

RESUMEN

ErbB-3 binding protein 1 (Ebp1) is a host protein which binds ErbB-3 receptor to induce signalling events for cell growth regulation. In addition, Ebp1 also interacts with ribonucleoprotein complexes. In recent times, Ebp1 was found to play an antagonistic role in viral infections caused by Influenza and Rinderpest viruses. In our present work we have tried to understand the role of Ebp1 in Chandipura virus (CHPV) infection. We have observed an induction in Ebp1 expression upon CHPV infection similar to other viruses. However, unlike other viruses an overexpressed Ebp1 only reduces viral protein expression, but does not affect its progeny formation. Additionally, this effect is being carried out in an indirect manner, as there is no interaction between Ebp1 and viral proteins. This is despite Ebp1's presence in viral inclusion bodies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Interacciones Huésped-Patógeno/genética , Neuronas/metabolismo , Proteínas de Unión al ARN/genética , Vesiculovirus/genética , Replicación Viral , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Regulación de la Expresión Génica , Humanos , Cuerpos de Inclusión Viral/química , Neuronas/virología , Plásmidos/química , Plásmidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Transfección , Células Vero , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/metabolismo , Ensayo de Placa Viral
6.
J Virol ; 93(15)2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31092575

RESUMEN

Oncolytic virotherapy represents a promising experimental anticancer strategy, based on the use of genetically modified viruses to selectively infect and kill cancer cells. Vesicular stomatitis virus (VSV) is a prototypic oncolytic virus (OV) that induces cancer cell death through activation of the apoptotic pathway, although intrinsic resistance to oncolysis is found in some cell lines and many primary tumors, as a consequence of residual innate immunity to the virus. In the effort to improve OV therapeutic efficacy, we previously demonstrated that different agents, including histone deacetylase inhibitors (HDIs), functioned as reversible chemical switches to dampen the innate antiviral response and improve the susceptibility of resistant cancer cells to VSV infection. In the present study, we demonstrated that the NAD+-dependent histone deacetylase SIRT1 (silent mating type information regulation 2 homolog 1) plays a key role in the permissivity of prostate cancer PC-3 cells to VSVΔM51 replication and oncolysis. HDI-mediated enhancement of VSVΔM51 infection and cancer cell killing directly correlated with a decrease of SIRT1 expression. Furthermore, pharmacological inhibition as well as silencing of SIRT1 by small interfering RNA (siRNA) was sufficient to sensitize PC-3 cells to VSVΔM51 infection, resulting in augmentation of virus replication and spread. Mechanistically, HDIs such as suberoylanilide hydroxamic acid (SAHA; Vorinostat) and resminostat upregulated the microRNA miR-34a that regulated the level of SIRT1. Taken together, our findings identify SIRT1 as a viral restriction factor that limits VSVΔM51 infection and oncolysis in prostate cancer cells.IMPORTANCE The use of nonpathogenic viruses to target and kill cancer cells is a promising strategy in cancer therapy. However, many types of human cancer are resistant to the oncolytic (cancer-killing) effects of virotherapy. In this study, we identify a host cellular protein, SIRT1, that contributes to the sensitivity of prostate cancer cells to infection by a prototypical oncolytic virus. Knockout of SIRT1 activity increases the sensitivity of prostate cancer cells to virus-mediated killing. At the molecular level, SIRT1 is controlled by a small microRNA termed miR-34a. Altogether, SIRT1 and/or miR-34a levels may serve as predictors of response to oncolytic-virus therapy.


Asunto(s)
Interacciones Microbiota-Huesped , Inmunidad Innata , Virus Oncolíticos/crecimiento & desarrollo , Sirtuina 1/metabolismo , Vesiculovirus/crecimiento & desarrollo , Replicación Viral , Humanos , Masculino , Virus Oncolíticos/inmunología , Células PC-3 , Vesiculovirus/inmunología
7.
Virology ; 532: 22-29, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30999160

RESUMEN

Priming of the viral glycoprotein (GP) by the cellular proteases cathepsin B and L (CatB, CatL) is believed to be essential for cell entry of filoviruses. However, pseudotyping systems that predominantly produce non-filamentous particles have frequently been used to prove this concept. Here, we report that GP-mediated entry of retroviral-, rhabdoviral and filoviral particles depends on CatB/CatL activity and that this effect is cell line-independent. Moreover, we show that the human cell line Calu-3, which expresses low amounts of CatL, is largely resistant to entry driven by diverse filovirus GPs. Finally, we demonstrate that Calu-3 cell entry mediated by certain filovirus GPs can be rescued upon directed expression of CatL or DC-SIGN. Our results identify Calu-3 cells as largely resistant to filovirus GP-driven entry and demonstrate that entry is limited at the stage of virion attachment and GP priming.


Asunto(s)
Catepsina L/genética , Moléculas de Adhesión Celular/genética , Ebolavirus/genética , Células Epiteliales/inmunología , Lectinas Tipo C/genética , Receptores de Superficie Celular/genética , Proteínas Virales/genética , Células A549 , Animales , Catepsina B/antagonistas & inhibidores , Catepsina B/genética , Catepsina B/inmunología , Catepsina B/metabolismo , Catepsina L/antagonistas & inhibidores , Catepsina L/inmunología , Catepsina L/metabolismo , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Chlorocebus aethiops , Inhibidores de Cisteína Proteinasa/farmacología , Dipéptidos/farmacología , Ebolavirus/crecimiento & desarrollo , Ebolavirus/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/virología , Regulación de la Expresión Génica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno/genética , Humanos , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Leucina/análogos & derivados , Leucina/farmacología , Marburgvirus/genética , Marburgvirus/crecimiento & desarrollo , Marburgvirus/metabolismo , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Células Vero , Vesiculovirus/genética , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/metabolismo , Proteínas Virales/metabolismo , Virión/genética , Virión/crecimiento & desarrollo , Virión/metabolismo , Internalización del Virus/efectos de los fármacos
8.
Virology ; 526: 32-37, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30336336

RESUMEN

Snakehead vesiculovirus (SHVV) is a new rhabdovirus isolated from diseased hybrid snakehead fish (Channa maculate ♀ x Channa argus ♂) and has caused serious economic losses in snakehead fish culture in China. To better understand the pathogenicity of SHVV, we developed a reverse genetics system for SHVV by using human and fish cells. In detail, human 293T cells were co-transfected with four plasmids encoding the full-length SHVV antigenomic RNA or the supporting proteins including nucleoprotein (N), phosphoprotein (P), and large polymerase (L), followed by the cultivation in Channel catfish ovary (CCO) cells. We also rescued a recombinant SHVV expressing enhanced green fluorescent protein (EGFP), which was inserted into the 3' non-coding region (NCR) of the glycoprotein (G) gene of SHVV. Our study provides a potential tool for unveiling the pathogenicity of SHVV and a template for the rescue of other fish viruses by using both human 293T and fish cells.


Asunto(s)
Enfermedades de los Peces/virología , Genética Inversa , Infecciones por Rhabdoviridae/virología , Vesiculovirus/genética , Animales , Células Cultivadas , ADN Complementario/genética , Femenino , Peces , Genes Virales/genética , Genoma Viral/genética , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Ovario/citología , ARN Viral/biosíntesis , ARN Viral/genética , Vesiculovirus/crecimiento & desarrollo
9.
Protein Expr Purif ; 153: 53-58, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30165247

RESUMEN

Recombinant interferon-α (rIFN-α) has been widely used for treating viral infections. However, the clinical efficacy of unmodified rIFN-α is limited due to small molecular size and rapid clearance from circulation. In this study we developed a novel strategy for half-life extension of porcine IFN-α (PoIFN-α) by fusion to the immunoglobulin (Ig)-binding C2 domain of streptococcal protein G (SPG). The coding sequences for PoIFN-α6 and SPG C2 domain, with a tobacco etch virus (TEV) protease recognition sequence introduced at the 5-end, were cloned into an elastin-like polypeptide (ELP) fusion expression vector and expressed as an ELP-PoIFNα-C2 fusion protein. After optimization of the conditions for soluble protein expression and purification, the fusion protein was purified to more than 90% purity by two rounds of inverse transition cycling (ITC) in the presence of 0.5% Triton X-100. After cleavage with self-aggregating peptide ELK-16-tagged tobacco etch virus protease, the protease was removed by quick centrifugation and PoIFNα-C2 protein was recovered by an additional round of ITC with 98% purity. Western blotting analysis showed that PoIFNα-C2 protein had the specific affinity for pig IgG binding. The antiviral assay showed that PoIFNα-C2 protein had potent antiviral activities against vesicular stomatitis virus and porcine pseudorabies virus. After single intravenous or subcutaneous injection into rats, PoIFNα-C2 protein showed 16- or 4-fold increase in serum half-life with significantly improved bioavailability.


Asunto(s)
Proteínas Bacterianas/farmacocinética , Herpesvirus Suido 1/efectos de los fármacos , Interferón-alfa/farmacocinética , Proteínas Recombinantes de Fusión/farmacocinética , Vesiculovirus/efectos de los fármacos , Secuencias de Aminoácidos , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Bioensayo , Disponibilidad Biológica , Línea Celular , Clonación Molecular , Elastina/genética , Elastina/metabolismo , Endopeptidasas/química , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Semivida , Herpesvirus Suido 1/crecimiento & desarrollo , Herpesvirus Suido 1/inmunología , Humanos , Interferón-alfa/genética , Interferón-alfa/inmunología , Péptidos/genética , Péptidos/metabolismo , Unión Proteica , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Porcinos , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/inmunología
10.
Mol Immunol ; 106: 46-52, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30576951

RESUMEN

The bovine IFN-ω (BoIFN-ω) multigene family is located on chromosome 8, which has 14 potential functional genes and 10 pseudogenes. After aligning 14 BoIFN-ω subtypes and assigning the most frequently occurring amino acids in each position, one artificial consensus BoIFN-ω (CoBoIFN-ω) gene was designed, optimized and synthesized. Then, CoBoIFN-ω was expressed in Pichia pastoris, which was demonstrated to have 3.94-fold and 14.3-fold higher antiviral activity against VSV on MDBK cells than that of BoIFN-ω24 and BoIFN-ω3, respectively. Besides this, CoBoIFN-ω was confirmed to have antiviral activity against VSV on BL, BT, PK-15 cells, and against BEV, BHV-1, BPIV3 on MDBK cells. Additionally, CoBoIFN-ω could bind with bovine type I IFN receptors, and then activate the promoters of NF-κB, ISRE and BoIFN-ß, and induce the transcription of ISGs and expression of Mx1 and NF-κB p65, which suggested CoBoIFN-ω exerts antiviral activity via activation of the JAK-STAT signaling pathway. Overall, this research on CoBoIFN-ω not only extends and improves consensus IFN research, but also reveals that CoBoIFN-ω has the potential to be used in the therapy of bovine viral diseases.


Asunto(s)
Antivirales , Herpesvirus Bovino 1/crecimiento & desarrollo , Interferón Tipo I , Virus de la Parainfluenza 3 Bovina/crecimiento & desarrollo , Pichia/metabolismo , Transducción de Señal , Vesiculovirus/crecimiento & desarrollo , Animales , Antivirales/inmunología , Antivirales/aislamiento & purificación , Antivirales/metabolismo , Antivirales/farmacología , Bovinos , Línea Celular , Cricetinae , Interferón Tipo I/biosíntesis , Interferón Tipo I/química , Interferón Tipo I/inmunología , Interferón Tipo I/farmacología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
11.
J Gen Virol ; 100(2): 278-288, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30566072

RESUMEN

A first step towards the development of a human immunodeficiency virus (HIV) animal model has been the identification and surmounting of species-specific barriers encountered by HIV along its replication cycle in cells from small animals. Serine incorporator proteins 3 (SERINC3) and 5 (SERINC5) were recently identified as restriction factors that reduce HIV-1 infectivity. Here, we compared the antiviral activity of SERINC3 and SERINC5 among mice, rats and rabbits, and their susceptibility to viral counteraction to their human counterparts. In the absence of viral antagonists, rodent and lagomorph SERINC3 and SERINC5 displayed anti-HIV activity in a similar range to human controls. Vesicular stomatitis virus G protein (VSV-G) pseudotyped virions were considerably less sensitive to restriction by all SERINC3/5 orthologs. Interestingly, HIV-1 Nef, murine leukemia virus (MLV) GlycoGag and equine infectious anemia virus (EIAV) S2 counteracted the antiviral activity of all SERINC3/5 orthologs with similar efficiency. Our results demonstrate that the antiviral activity of SERINC3/5 proteins is conserved in rodents and rabbits, and can be overcome by all three previously reported viral antagonists.


Asunto(s)
VIH-1/crecimiento & desarrollo , VIH-1/inmunología , Interacciones Huésped-Patógeno , Factores Inmunológicos/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Vectores Genéticos , Ratones , Conejos , Ratas , Vesiculovirus/genética , Vesiculovirus/crecimiento & desarrollo
12.
Viral Immunol ; 31(10): 676-682, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30265587

RESUMEN

microRNAs have been reported to play crucial roles in various biological processes, including cell proliferation, apoptosis, tumor genesis, and viral infections. miR-26b has been found to be involved in the pathogenesis of multiple tumors, however, little is known about the role it plays in innate immune responses. In this study, we report that miR-26b is able to induce type-I interferon (IFN) expression, which was supported by both quantitative real time polymerase chain reaction and luciferase reporter assays. Conversely, production of IFN was reduced upon inhibition of miR-26b. Sequentially, ectopic expression of miR-26b led to upregulated expression of STAT1 and IFN-stimulated genes (ISGs). Furthermore, overexpression of miR-26b repressed the replication of vesicular stomatitis virus (VSV) and Sendai virus (SeV). In turn, IFN was able to induce the expression of miR-26b in a time-dependent manner. In all, we found that miR-26b could inhibit VSV replication through upregulation of type-I IFNs and ISGs and could in turn be upregulated by IFNs.


Asunto(s)
Inmunidad Innata , Interferón Tipo I/metabolismo , MicroARNs/metabolismo , Virus Sendai/inmunología , Transducción de Señal , Vesiculovirus/inmunología , Replicación Viral , Perfilación de la Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Factores Inmunológicos/biosíntesis , Factores Inmunológicos/genética , Luciferasas/análisis , Luciferasas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Virus Sendai/crecimiento & desarrollo , Vesiculovirus/crecimiento & desarrollo
13.
Virology ; 522: 244-259, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30055515

RESUMEN

Vesicular stomatitis virus (VSV) shows potential for targeting and killing cancer cells, but can be dangerous in the brain due to its neurotropic glycoprotein. Here we test a chimeric virus in which the VSV glycoprotein is replaced with the Chikungunya polyprotein E3-E2-6K-E1 (VSVΔG-CHIKV). Control mice with brain tumors survived a mean of 40 days after tumor implant. VSVΔG-CHIKV selectively infected and eliminated the tumor, and extended survival substantially in all tumor-bearing mice to over 100 days. VSVΔG-CHIKV also targeted intracranial primary patient derived melanoma xenografts. Virus injected into one melanoma spread to other melanomas within the same brain with little detectable infection of normal cells. Intravenous VSVΔG-CHIKV infected tumor cells but not normal tissue. In immunocompetent mice, VSVΔG-CHIKV selectively infected mouse melanoma cells within the brain. These data suggest VSVΔG-CHIKV can target and destroy brain tumors in multiple animal models without the neurotropism associated with the wild type VSV glycoprotein.


Asunto(s)
Neoplasias Encefálicas/terapia , Virus Chikungunya/crecimiento & desarrollo , Virus Chikungunya/genética , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/genética , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/genética , Animales , Modelos Animales de Enfermedad , Xenoinjertos , Melanoma/terapia , Ratones , Trasplante de Neoplasias , Viroterapia Oncolítica , Recombinación Genética , Análisis de Supervivencia , Resultado del Tratamiento
14.
Pharmacology ; 101(3-4): 148-155, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29275416

RESUMEN

When the Kampo medicine, Hochuekkito (Hochu), was administered to normal mice for 2 weeks, influenza virus titer was reduced. The mechanism of action of Hochu was examined using the plaque assay method. It was suggested that Hochu may either obstruct the first stage of the infection process (adsorption and entry) or may directly target viral particles. Using the plaque assay method, these 2 modes of action could not be differentiated. Virus RNA in the infected cell was verified by quantitative real-time polymerase chain reaction. An equal inhibition effect was obtained when Hochu was preprocessed for normal cells and when they were made to act simultaneously with virus adsorption. The viral load at the cell surface following UV irradiation was higher in the Hochu-administered group as compared with that of the control. Moreover, the affinity of Hochu for the influenza virus was hundred times higher than its affinity for the host cell. The effect of entry obstruction by Hochu was observed via image analysis, where the amount of virus nucleocapsid protein (NP) invading the cell was visualized with FITC-labeled NP antibody. Hochu does not seem to have an effect on nucleic acid synthesis, viral release from infected cells, and on the subsequent second round of infection. In conclusion, Hochu binds to viral particles and forms complexes that can obstruct the entry of influenza virus into cells.


Asunto(s)
Antivirales/uso terapéutico , Medicamentos Herbarios Chinos/uso terapéutico , Subtipo H1N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Animales , Línea Celular , Perros , Humanos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H1N1 del Virus de la Influenza A/fisiología , Células de Riñón Canino Madin Darby , Masculino , Medicina Kampo , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/virología , Fitoterapia , ARN Viral/análisis , Vesiculovirus/crecimiento & desarrollo , Ensayo de Placa Viral , Internalización del Virus/efectos de los fármacos
15.
Biometals ; 31(1): 81-89, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29209895

RESUMEN

Here we report on the results obtained from an antiviral screening, including herpes simplex virus, vaccinia virus, vesicular stomatitis virus, Coxsackie B4 virus or respiratory syncytial virus, parainfluenza-3 virus, reovirus-1 and Punta Toro virus, of three 2-hydroxy-3-methoxyphenyl acylhydrazone compounds in three cell lines (i.e. human embryonic lung fibroblast cells, human cervix carcinoma cells, and African Green monkey kidney cells). Interesting antiviral EC50 values are obtained against herpes simplex virus-1 and vaccinia virus. The biological activity of acylhydrazones is often attributed to their metal coordinating abilities, so potentiometric and microcalorimetric studies are here discussed to unravel the behavior of the three 2-hydroxy-3-methoxyphenyl compounds in solution. It is worth of note that the acylhydrazone with the higher affinity for Cu(II) ions shows the best antiviral activity against herpes simplex and vaccinia virus (EC50 ~ 1.5 µM, minimal cytotoxic concentration = 60 µM, selectivity index = 40).


Asunto(s)
Antivirales/farmacología , Quelantes/farmacología , Hidrazonas/farmacología , Simplexvirus/efectos de los fármacos , Virus Vaccinia/efectos de los fármacos , Animales , Antivirales/síntesis química , Antivirales/metabolismo , Línea Celular , Línea Celular Tumoral , Quelantes/síntesis química , Quelantes/metabolismo , Chlorocebus aethiops , Cobre/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Humanos , Hidrazonas/síntesis química , Hidrazonas/metabolismo , Concentración 50 Inhibidora , Magnesio/metabolismo , Manganeso/metabolismo , Orthoreovirus de los Mamíferos/efectos de los fármacos , Orthoreovirus de los Mamíferos/crecimiento & desarrollo , Orthoreovirus de los Mamíferos/metabolismo , Virus de la Parainfluenza 3 Humana/efectos de los fármacos , Virus de la Parainfluenza 3 Humana/crecimiento & desarrollo , Virus de la Parainfluenza 3 Humana/metabolismo , Phlebovirus/efectos de los fármacos , Phlebovirus/crecimiento & desarrollo , Phlebovirus/metabolismo , Virus Sincitiales Respiratorios/efectos de los fármacos , Virus Sincitiales Respiratorios/crecimiento & desarrollo , Virus Sincitiales Respiratorios/metabolismo , Simplexvirus/crecimiento & desarrollo , Simplexvirus/metabolismo , Virus Vaccinia/crecimiento & desarrollo , Virus Vaccinia/metabolismo , Células Vero , Vesiculovirus/efectos de los fármacos , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/metabolismo
16.
J Infect Dis ; 217(5): 721-730, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29228368

RESUMEN

Cells latently infected with human immunodeficiency virus (HIV) evade immune- and drug-mediated clearance. These cells harbor intracellular signaling defects, including impairment of the antiviral type I interferon response. Such defects have also been observed in several cancers and have been exploited for the development of therapeutic oncolytic viruses, including the recombinant Maraba virus (MG1). We therefore hypothesized that MG1 would infect and eliminate cells latently infected with HIV-1, while sparing healthy uninfected cells. Preferential infection and elimination by MG1 was first demonstrated in cell lines latently infected with HIV-1. Following this, a reduction in HIV-1 DNA and inducible HIV-1 replication was observed following MG1 infection of latently infected, resting CD4+ T cells generated using an in vitro model of latency. Last, MG1 infection resulted in a reduction in HIV-1 DNA and inducible HIV-1 replication in memory CD4+ T cells isolated from effectively treated, HIV-1-infected individuals. Our results therefore highlight a novel approach to eliminate the latent HIV-1 reservoir.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Infecciones por VIH/virología , VIH-1/fisiología , Virus Oncolíticos/crecimiento & desarrollo , Vesiculovirus/crecimiento & desarrollo , Latencia del Virus , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Infecciones por VIH/terapia , Humanos
17.
Virus Res ; 244: 199-207, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29155034

RESUMEN

2', 5'-Oligoadenylate synthetase-lilke (OASL) protein is an atypical oligoadenylate synthetase (OAS) family member, which possesses antiviral activity but lacks 2', 5'-oligoadenylate synthetase activity. Here, a novel variant of porcine OASL (pOASL2) was identified through RT-PCR amplification. This gene is distinguishable from the previously described wild-type porcine OASL (pOASL1). The gene appears to be derived from a truncation of exon 4 plus 8 nucleotides of exon 5 with a premature termination, measuring only 633 bp in length, although its position corresponds to that of pOASL1. Given this novel gene appears to be a variant of pOASL, we assayed for antiviral activity of the protein. We demonstrated that pOASL2 could inhibit Japanese encephalitis virus (JEV) proliferation as well as pOASL1 in a transient overexpression assay of pOASL1 and pOASL2 in PK-15 and Vero cells. In addition to JEV, pOASL1 and pOASL2 also decreased the proliferations of Porcine reproductive and respiratory syndrome virus (PRRSV) and vesicular stomatitis virus (VSV), but did not exhibit antiviral activity against pseudorabies virus (PRV). Structural analysis showed that the pOASL2 gene retained only the first three exons at the 5'-. To investigate the role of the αN4 helix in pOASL in antiviral responses like that in hOASL, we mutated key residues in the anchor domain of the αN4 helix in pOASL2, based on the domain's location in hOASL. However, the antiviral activity of pOASL2 was not affected. Thus, the αN4 helix of pOASL likely does not play a significant role in its antiviral activity. In conclusion, pOASL2 acts as a new splice isoform of pOASL that plays a role in resistance to infection of several kinds of RNA viruses.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/farmacología , Empalme Alternativo , Antivirales/farmacología , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Virus del Síndrome Respiratorio y Reproductivo Porcino/efectos de los fármacos , Vesiculovirus/efectos de los fármacos , 2',5'-Oligoadenilato Sintetasa/biosíntesis , 2',5'-Oligoadenilato Sintetasa/química , 2',5'-Oligoadenilato Sintetasa/genética , Secuencia de Aminoácidos , Animales , Antivirales/química , Línea Celular , Chlorocebus aethiops , Virus de la Encefalitis Japonesa (Especie)/genética , Virus de la Encefalitis Japonesa (Especie)/crecimiento & desarrollo , Virus de la Encefalitis Japonesa (Especie)/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Células Epiteliales/virología , Exones , Herpesvirus Suido 1/efectos de los fármacos , Herpesvirus Suido 1/genética , Herpesvirus Suido 1/crecimiento & desarrollo , Herpesvirus Suido 1/metabolismo , Isoenzimas/biosíntesis , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/farmacología , Riñón/efectos de los fármacos , Riñón/patología , Riñón/virología , Sistemas de Lectura Abierta , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Virus del Síndrome Respiratorio y Reproductivo Porcino/crecimiento & desarrollo , Virus del Síndrome Respiratorio y Reproductivo Porcino/metabolismo , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Porcinos , Células Vero , Vesiculovirus/genética , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/metabolismo
18.
J Virol ; 91(15)2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28539437

RESUMEN

Ebolaviruses have a surface glycoprotein (GP1,2) that is required for virus attachment and entry into cells. Mutations affecting GP1,2 functions can alter virus growth properties. We generated a recombinant vesicular stomatitis virus encoding Ebola virus Makona variant GP1,2 (rVSV-MAK-GP) and observed emergence of a T544I mutation in the Makona GP1,2 gene during tissue culture passage in certain cell lines. The T544I mutation emerged within two passages when VSV-MAK-GP was grown on Vero E6, Vero, and BS-C-1 cells but not when it was passaged on Huh7 and HepG2 cells. The mutation led to a marked increase in virus growth kinetics and conferred a robust growth advantage over wild-type rVSV-MAK-GP on Vero E6 cells. Analysis of complete viral genomes collected from patients in western Africa indicated that this mutation was not found in Ebola virus clinical samples. However, we observed the emergence of T544I during serial passage of various Ebola Makona isolates on Vero E6 cells. Three independent isolates showed emergence of T544I from undetectable levels in nonpassaged virus or virus passaged once to frequencies of greater than 60% within a single passage, consistent with it being a tissue culture adaptation. Intriguingly, T544I is not found in any Sudan, Bundibugyo, or Tai Forest ebolavirus sequences. Furthermore, T544I did not emerge when we serially passaged recombinant VSV encoding GP1,2 from these ebolaviruses. This report provides experimental evidence that the spontaneous mutation T544I is a tissue culture adaptation in certain cell lines and that it may be unique for the species Zaire ebolavirusIMPORTANCE The Ebola virus (Zaire) species is the most lethal species of all ebolaviruses in terms of mortality rate and number of deaths. Understanding how the Ebola virus surface glycoprotein functions to facilitate entry in cells is an area of intense research. Recently, three groups independently identified a polymorphism in the Ebola glycoprotein (I544) that enhanced virus entry, but they did not agree in their conclusions regarding its impact on pathogenesis. Our findings here address the origins of this polymorphism and provide experimental evidence showing that it is the result of a spontaneous mutation (T544I) specific to tissue culture conditions, suggesting that it has no role in pathogenesis. We further show that this mutation may be unique to the species Zaire ebolavirus, as it does not occur in Sudan, Bundibugyo, and Tai Forest ebolaviruses. Understanding the mechanism behind this mutation can provide insight into functional differences that exist in culture conditions and among ebolavirus glycoproteins.


Asunto(s)
Ebolavirus/fisiología , Proteínas Mutantes/genética , Mutación Missense , Selección Genética , Proteínas del Envoltorio Viral/genética , Internalización del Virus , Adaptación Biológica , Sustitución de Aminoácidos , Animales , Línea Celular , Análisis Mutacional de ADN , Ebolavirus/genética , Ebolavirus/crecimiento & desarrollo , Genoma Viral , Humanos , Recombinación Genética , Genética Inversa , Análisis de Secuencia de ADN , Pase Seriado , Vesiculovirus/genética , Vesiculovirus/crecimiento & desarrollo , Cultivo de Virus
19.
Retrovirology ; 14(1): 21, 2017 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-28330477

RESUMEN

BACKGROUND: Human endogenous retroviruses (HERVs) are genomic sequences of retroviral origin which were believed to be integrated into germline chromosomes millions of years ago and account for nearly 8% of the human genome. Although mostly defective and inactive, some of the HERVs may be activated under certain physiological and pathological conditions. While no drugs are designed specifically targeting HERVs, there are a panel of antiretroviral drugs designed against the human immunodeficiency virus and approved by the Federal Drug Administration (FDA). RESULTS: We determined if these antiretroviral drugs may also be effective in inhibiting HERVs. We constructed a plasmid with consensus HERV-K sequence for testing the effect of antiretroviral drugs on HERV-K. We first determined the effects of nucleoside and non-nucleotide reverse transcriptase (RT) inhibitors on HERV-K by product enhanced reverse transcription assay. We found that all RT inhibitors could significantly inhibit HERV-K RT activity. To determine the effects of antiretroviral drugs on HERV-K infection and viral production, we pseudotyped HERV-K with VSV-G and used the pseudotyped HERV-K virus to infect HeLa cells. HERV-K production was measured by quantitative real time polymerase chain reaction. We found that RT inhibitors Abacavir and Zidovudine, and integrase inhibitor Raltegravir could effectively block HERV-K infection and production. However, protease inhibitors were not as effective as RT and integrase inhibitors. CONCLUSIONS: In summary, we identified several FDA approved antiretroviral drugs that can effectively inhibit HERV-K. These antiretrovirals may open new prospects for studying HERV-K pathophysiology and potentially for exploring treatment of diseases in which HERV-K has been implicated.


Asunto(s)
Antirretrovirales/farmacología , Retrovirus Endógenos/efectos de los fármacos , Células HeLa , Humanos , Pruebas de Sensibilidad Microbiana , Reacción en Cadena en Tiempo Real de la Polimerasa , Recombinación Genética , Vesiculovirus/genética , Vesiculovirus/crecimiento & desarrollo
20.
Proc Natl Acad Sci U S A ; 114(1): E95-E104, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-27980033

RESUMEN

The brain has a tightly regulated environment that protects neurons and limits inflammation, designated "immune privilege." However, there is not an absolute lack of an immune response. We tested the ability of the brain to initiate an innate immune response to a virus, which was directly injected into the brain parenchyma, and to determine whether this response could limit viral spread. We injected vesicular stomatitis virus (VSV), a transsynaptic tracer, or naturally occurring VSV-derived defective interfering particles (DIPs), into the caudate-putamen (CP) and scored for an innate immune response and inhibition of virus spread. We found that the brain parenchyma has a functional type I interferon (IFN) response that can limit VSV spread at both the inoculation site and among synaptically connected neurons. Furthermore, we characterized the response of microglia to VSV infection and found that infected microglia produced type I IFN and uninfected microglia induced an innate immune response following virus injection.


Asunto(s)
Encéfalo/inmunología , Inmunidad Innata/inmunología , Interferón Tipo I/inmunología , Tejido Parenquimatoso/inmunología , Vesiculovirus/inmunología , Animales , Encéfalo/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Tejido Parenquimatoso/virología , Estomatitis Vesicular/inmunología , Estomatitis Vesicular/virología , Vesiculovirus/crecimiento & desarrollo , Replicación Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...