Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Gen Virol ; 97(9): 2096-2103, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27439314

RESUMEN

Avian bornaviruses are the causative agents of proventricular dilatation disease (PDD), a widely distributed and often fatal disease in captive psittacines. Because neither specific prevention measures nor therapies against PDD and bornavirus infections are currently available, new antiviral strategies are required to improve animal health. We show here that the nucleoside analogue ribavirin inhibited bornavirus activity in a polymerase reconstitution assay and reduced viral load in avian cell lines infected with two different parrot bornaviruses. Furthermore, we observed that ribavirin enhanced type I IFN signalling in avian cells. Combined treatment of avian bornavirus-infected cells with ribavirin and recombinant IFN-α strongly enhanced the antiviral efficiency compared to either drug alone. The combined use of ribavirin and type I IFN might represent a promising new strategy for therapeutic treatment of captive parrots persistently infected with avian bornaviruses.


Asunto(s)
Antivirales/farmacología , Bornaviridae/efectos de los fármacos , Interferón-alfa/farmacología , Ribavirina/farmacología , Animales , Línea Celular , Pruebas de Sensibilidad Microbiana , Loros , Carga Viral
2.
PLoS Pathog ; 11(11): e1005264, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26588843

RESUMEN

Sensing viruses by pattern recognition receptors (PRR) triggers the innate immune system of the host cell and activates immune signaling cascades such as the RIG-I/IRF3 pathway. Mitochondrial antiviral-signaling protein (MAVS, also known as IPS-1, Cardif, and VISA) is the crucial adaptor protein of this pathway localized on mitochondria, peroxisomes and mitochondria-associated membranes of the endoplasmic reticulum. Activation of MAVS leads to the production of type I and type III interferons (IFN) as well as IFN stimulated genes (ISGs). To refine the role of MAVS subcellular localization for the induction of type I and III IFN responses in hepatocytes and its counteraction by the hepatitis C virus (HCV), we generated various functional and genetic knock-out cell systems that were reconstituted to express mitochondrial (mito) or peroxisomal (pex) MAVS, exclusively. Upon infection with diverse RNA viruses we found that cells exclusively expressing pexMAVS mounted sustained expression of type I and III IFNs to levels comparable to cells exclusively expressing mitoMAVS. To determine whether viral counteraction of MAVS is affected by its subcellular localization we employed infection of cells with HCV, a major causative agent of chronic liver disease with a high propensity to establish persistence. This virus efficiently cleaves MAVS via a viral protease residing in its nonstructural protein 3 (NS3) and this strategy is thought to contribute to the high persistence of this virus. We found that both mito- and pexMAVS were efficiently cleaved by NS3 and this cleavage was required to suppress activation of the IFN response. Taken together, our findings indicate comparable activation of the IFN response by pex- and mitoMAVS in hepatocytes and efficient counteraction of both MAVS species by the HCV NS3 protease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Retículo Endoplásmico/metabolismo , Hepacivirus , Interferones/metabolismo , Mitocondrias/virología , Peroxisomas/virología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular , Retículo Endoplásmico/virología , Hepatocitos/metabolismo , Humanos , Ratones , Mitocondrias/metabolismo , Peroxisomas/metabolismo , Proteínas no Estructurales Virales/metabolismo
3.
PLoS Pathog ; 11(12): e1005345, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26720415

RESUMEN

Dengue virus (DENV) is the most common mosquito-transmitted virus infecting ~390 million people worldwide. In spite of this high medical relevance, neither a vaccine nor antiviral therapy is currently available. DENV elicits a strong interferon (IFN) response in infected cells, but at the same time actively counteracts IFN production and signaling. Although the kinetics of activation of this innate antiviral defense and the timing of viral counteraction critically determine the magnitude of infection and thus disease, quantitative and kinetic analyses are lacking and it remains poorly understood how DENV spreads in IFN-competent cell systems. To dissect the dynamics of replication versus antiviral defense at the single cell level, we generated a fully viable reporter DENV and host cells with authentic reporters for IFN-stimulated antiviral genes. We find that IFN controls DENV infection in a kinetically determined manner that at the single cell level is highly heterogeneous and stochastic. Even at high-dose, IFN does not fully protect all cells in the culture and, therefore, viral spread occurs even in the face of antiviral protection of naïve cells by IFN. By contrast, a vaccine candidate DENV mutant, which lacks 2'-O-methylation of viral RNA is profoundly attenuated in IFN-competent cells. Through mathematical modeling of time-resolved data and validation experiments we show that the primary determinant for attenuation is the accelerated kinetics of IFN production. This rapid induction triggered by mutant DENV precedes establishment of IFN-resistance in infected cells, thus causing a massive reduction of virus production rate. In contrast, accelerated protection of naïve cells by paracrine IFN action has negligible impact. In conclusion, these results show that attenuation of the 2'-O-methylation DENV mutant is primarily determined by kinetics of autocrine IFN action on infected cells.


Asunto(s)
Vacunas contra el Dengue/inmunología , Virus del Dengue/inmunología , Dengue/inmunología , Interferones/inmunología , Modelos Teóricos , Línea Celular , Supervivencia Celular , Vacunas contra el Dengue/genética , Virus del Dengue/genética , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Metilación , ARN Viral/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
J Virol ; 88(5): 2835-43, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24371053

RESUMEN

Interferons (IFNs) are essential components of the antiviral defense system of vertebrates. In mammals, functional receptors for type III IFN (lambda interferon [IFN-λ]) are found mainly on epithelial cells, and IFN-λ was demonstrated to play a crucial role in limiting viral infections of mucosal surfaces. To determine whether IFN-λ plays a similar role in birds, we produced recombinant chicken IFN-λ (chIFN-λ) and we used the replication-competent retroviral RCAS vector system to generate mosaic-transgenic chicken embryos that constitutively express chIFN-λ. We could demonstrate that chIFN-λ markedly inhibited replication of various virus strains, including highly pathogenic influenza A viruses, in ovo and in vivo, as well as in epithelium-rich tissue and cell culture systems. In contrast, chicken fibroblasts responded poorly to chIFN-λ. When applied in vivo to 3-week-old chickens, recombinant chIFN-λ strongly induced the IFN-responsive Mx gene in epithelium-rich organs, such as lungs, tracheas, and intestinal tracts. Correspondingly, these organs were found to express high transcript levels of the putative chIFN-λ receptor alpha chain (chIL28RA) gene. Transfection of chicken fibroblasts with a chIL28RA expression construct rendered these cells responsive to chIFN-λ treatment, indicating that receptor expression determines cell type specificity of IFN-λ action in chickens. Surprisingly, mosaic-transgenic chickens perished soon after hatching, demonstrating a detrimental effect of constitutive chIFN-λ expression. Our data highlight fundamental similarities between the IFN-λ systems of mammals and birds and suggest that type III IFN might play a role in defending mucosal surfaces against viral intruders in most if not all vertebrates.


Asunto(s)
Antivirales/farmacología , Interferones/farmacología , Proteínas Recombinantes/farmacología , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Línea Celular , Embrión de Pollo , Pollos , Resistencia a la Enfermedad/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Genes Letales , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/patogenicidad , Gripe Aviar/tratamiento farmacológico , Gripe Aviar/virología , Interferones/genética , Interferones/metabolismo , Datos de Secuencia Molecular , Proteínas de Resistencia a Mixovirus/genética , Proteínas de Resistencia a Mixovirus/metabolismo , Receptores de Citocinas/química , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Técnicas de Cultivo de Tejidos , Transcripción Genética/efectos de los fármacos , Replicación Viral/efectos de los fármacos
5.
J Hepatol ; 59(6): 1331-41, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23933585

RESUMEN

Infections with the hepatitis C virus (HCV) are a major cause of chronic liver disease. While the acute phase of infection is mostly asymptomatic, this virus has the high propensity to establish persistence and in the course of one to several decades liver disease can develop. HCV is a paradigm for the complex interplay between the interferon (IFN) system and viral countermeasures. The virus induces an IFN response within the infected cell and is rather sensitive against the antiviral state triggered by IFNs, yet in most cases HCV persists. Numerous IFN-stimulated genes (ISGs) have been reported to suppress HCV replication, but in only a few cases we begin to understand the molecular mechanisms underlying antiviral activity. It is becoming increasingly clear that blockage of viral replication is mediated by the concerted action of multiple ISGs that target different steps of the HCV replication cycle. This review briefly summarizes the activation of the IFN system by HCV and then focuses on ISGs targeting the HCV replication cycle and their possible mode of action.


Asunto(s)
Hepacivirus/fisiología , Interferones/fisiología , 2',5'-Oligoadenilato Sintetasa/fisiología , Antígenos de Diferenciación/fisiología , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/fisiología , Hepacivirus/aislamiento & purificación , Humanos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Proteínas/fisiología , Receptores Inmunológicos , Receptores Toll-Like/fisiología , Replicación Viral , eIF-2 Quinasa/fisiología
6.
Hum Gene Ther ; 23(5): 492-507, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22171602

RESUMEN

Libraries based on the insertion of random peptide ligands into the capsid of adeno-associated virus type 2 (AAV2) have been widely used to improve the efficiency and selectivity of the AAV vector system. However, so far only libraries of 7-mer peptide ligands have been inserted at one well-characterized capsid position. Here, we expanded the combinatorial AAV2 display system to a panel of novel AAV libraries, displaying peptides of 5, 7, 12, 19, or 26 amino acids in length at capsid position 588 or displaying 7-mer peptides at position 453, the most prominently exposed region of the viral capsid. Library selections on two unrelated cell types-human coronary artery endothelial cells and rat cardiomyoblasts-revealed the isolation of cell type-characteristic peptides of different lengths mediating strongly improved target-cell transduction, except for the 26-mer peptide ligands. Characterization of vector selectivity by transduction of nontarget cells and comparative gene-transduction analysis using a panel of 44 human tumor cell lines revealed that insertion of different-length peptides allows targeting of distinct cellular receptors for cell entry with similar efficiency, but with different selectivity. The application of such novel AAV2 libraries broadens the spectrum of targetable receptors by capsid-modified AAV vectors and provides the opportunity to choose the best suited targeting ligand for a certain application from a number of different candidates.


Asunto(s)
Proteínas de la Cápside/genética , Terapia Genética/métodos , Biblioteca de Péptidos , Animales , Sitios de Unión , Proteínas de la Cápside/metabolismo , Línea Celular Tumoral , Dependovirus , Células Endoteliales , Vectores Genéticos , Humanos , Mutagénesis Insercional , Mioblastos Cardíacos , Neoplasias/metabolismo , Péptidos/genética , Péptidos/metabolismo , Unión Proteica , Ratas , Transducción Genética
7.
J Virol ; 85(16): 8307-15, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21632756

RESUMEN

The type I interferon (IFN) system plays an important role in antiviral defense against influenza A viruses (FLUAV), which are natural chicken pathogens. Studies of mice identified the Mx1 protein as a key effector molecule of the IFN-induced antiviral state against FLUAV. Chicken Mx genes are highly polymorphic, and recent studies suggested that an Asn/Ser polymorphism at amino acid position 631 determines the antiviral activity of the chicken Mx protein. By employing chicken embryo fibroblasts with defined Mx-631 polymorphisms and retroviral vectors for the expression of Mx isoforms in chicken cells and embryonated eggs, we show here that neither the 631Asn nor the 631Ser variant of chicken Mx was able to confer antiviral protection against several lowly and highly pathogenic FLUAV strains. Using a short interfering RNA (siRNA)-mediated knockdown approach, we noted that the antiviral effect of type I IFN in chicken cells was not dependent on Mx, suggesting that some other IFN-induced factors must contribute to the inhibition of FLUAV in chicken cells. Finally, we found that both isoforms of chicken Mx protein appear to lack GTPase activity, which might explain the observed lack of antiviral activity.


Asunto(s)
Pollos/inmunología , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Virus de la Influenza A/inmunología , Gripe Aviar/inmunología , Interferón Tipo I/inmunología , Animales , Células Cultivadas , Embrión de Pollo , Pollos/genética , Pollos/virología , Técnica del Anticuerpo Fluorescente , GTP Fosfohidrolasas/metabolismo , Proteínas de Resistencia a Mixovirus , Polimorfismo de Nucleótido Simple , Isoformas de Proteínas , Interferencia de ARN , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
J Virol ; 85(15): 7730-41, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21613402

RESUMEN

From infection studies with cultured chicken cells and experimental mammalian hosts, it is well known that influenza viruses use the nonstructural protein 1 (NS1) to suppress the synthesis of interferon (IFN). However, our current knowledge regarding the in vivo role of virus-encoded NS1 in chickens is much more limited. Here, we report that highly pathogenic avian influenza viruses of subtypes H5N1 and H7N7 lacking fully functional NS1 genes were attenuated in 5-week-old chickens. Surprisingly, in diseased birds infected with NS1 mutants, the IFN levels were not higher than in diseased birds infected with wild-type virus, suggesting that NS1 cannot suppress IFN gene expression in at least one cell population of infected chickens that produces large amounts of the cytokine in vivo. To address the question of why influenza viruses are highly pathogenic in chickens although they strongly activate the innate immune system, we determined whether recombinant chicken alpha interferon (IFN-α) can inhibit the growth of highly pathogenic avian influenza viruses in cultured chicken cells and whether it can ameliorate virus-induced disease in 5-week-old birds. We found that IFN treatment failed to confer substantial protection against challenge with highly pathogenic viruses, although it was effective against viruses with low pathogenic potential. Taken together, our data demonstrate that preventing the synthesis of IFN is not the primary role of the viral NS1 protein during infection of chickens. Our results further suggest that virus-induced IFN does not contribute substantially to resistance of chickens against highly pathogenic influenza viruses.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/fisiología , Subtipo H7N7 del Virus de la Influenza A/fisiología , Gripe Aviar/metabolismo , Interferones/biosíntesis , Animales , Secuencia de Bases , Pollos , Cartilla de ADN , Inmunohistoquímica , Gripe Aviar/virología , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas no Estructurales Virales/fisiología
9.
Viruses ; 2(4): 927-938, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21994661

RESUMEN

Like other pathogens that readily persist in animal hosts, members of the Bornaviridae family have evolved effective mechanisms to evade the innate immune response. The prototype of this virus family, Borna disease virus employs an unusual replication strategy that removes the triphosphates from the 5' termini of the viral RNA genome. This strategy allows the virus to avoid activation of RIG-I and other innate immune response receptors in infected cells. Here we determined whether the newly discovered avian bornaviruses (ABV) might use a similar strategy to evade the interferon response. We found that de novo infection of QM7 and CEC32 quail cells with two different ABV strains was efficiently inhibited by exogenous chicken IFN-α. IFN-α also reduced the viral load in QM7 and CEC32 cells persistently infected with both ABV strains, suggesting that ABV is highly sensitive to type I IFN. Although quail cells persistently infected with ABV contained high levels of viral RNA, the supernatants of infected cultures did not contain detectable levels of biologically active type I IFN. RNA from cells infected with ABV failed to induce IFN-ß synthesis if transfected into human cells. Furthermore, genomic RNA of ABV was susceptible to 5'-monophosphate-specific RNase, suggesting that it lacks 5'-triphospates like BDV. These results indicate that bornaviruses of mammals and birds use similar strategies to evade the host immune response.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA