Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Gen Virol ; 97(12): 3161-3173, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27692048

RESUMEN

Newcastle disease virus, a prototype avian paramyxovirus serotype 1 (APMV-1), causes economically devastating disease in avian species around the world. Newcastle disease is enzootic in Pakistan and recurrent outbreaks are frequent in multiple avian species even after continuous and extensive use of vaccines. A number of APMV-1 and pigeon paramyxovirus serotype 1 (PPMV-1) strains have been isolated and genetically characterized in recent years. However, the impact of recently characterized wild bird-origin APMVs in domestic poultry, and the potency of routinely used vaccines against these novel and genetically diverse viruses remain unknown. Here, we applied next-generation sequencing for unbiased complete genome characterization of APMV-1 and PPMV-1 strains isolated from clinically diseased peacocks (Pavocristatus) and pigeons (Columbalivia), respectively. Global phylodynamics and evolutionary analysis demonstrates Pigeon/MZS-UVAS-Pak/2014 is clustered into lineage 4 (or genotype VI) and Peacock/MZS-UVAS-Pak/2014 into lineage 5 (or genotype VII). The genomes of both isolates encoded for polybasic residues (112RRQKR↓F117) at the fusion protein cleavage motif along with a number of important substitutions in the surface glycoproteins compared with the vaccine strains. Clinicopathological and immunological investigations in domesticated chickens indicate that these isolates can potentially transmit between tested avian species, can cause systemic infections, and can induce antibodies that are unable to prevent virus shedding. Collectively, the data from these genomic and biological assessments highlight the potential of wild birds in transmitting APMVs to domesticated chickens. The study also demonstrates that the current vaccine regimens are incapable of providing complete protection against wild bird-origin APMVs and PPMVs.


Asunto(s)
Enfermedad de Newcastle/virología , Virus de la Enfermedad de Newcastle/inmunología , Enfermedades de las Aves de Corral/virología , Animales , Animales Salvajes/virología , Pollos/virología , Columbidae/virología , Genoma Viral , Genotipo , Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/patogenicidad , Virus de la Enfermedad de Newcastle/fisiología , Filogenia , Enfermedades de las Aves de Corral/inmunología , Proteínas Virales de Fusión/administración & dosificación , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología , Virulencia
2.
Proc Natl Acad Sci U S A ; 109(1): 279-84, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22184245

RESUMEN

Individual microRNAs (miRNAs) are rapidly down-regulated during conditions of cellular activation and infection, but factors mediating miRNA turnover are poorly understood. Infection of mouse cells with murine cytomegalovirus (MCMV) induces the rapid down-regulation of an antiviral cellular miRNA, miR-27. Here, we identify a transcript produced by MCMV that binds to miR-27 and mediates its degradation. UV-crosslinking and high-throughput sequencing [CRAC (UV-crosslinking and analysis of cDNA)] identified MCMV RNA segments associated with the miRNA-binding protein Argonaute 2 (Ago2). A cluster of hits mapped to a predicted miR-27-binding site in the 3'UTR of the previously uncharacterized ORF, m169. The expression kinetics of the m169 transcript correlated with degradation of miR-27 during infection, and m169 expression inhibited miR-27 functional activity in a reporter assay. siRNA knockdown of m169 demonstrated its requirement for miR-27 degradation following infection and did not affect other host miRNAs. Substitution of the miR-27-binding site in m169 to create complementarity to a different cellular miRNA, miR-24, resulted in down-regulation of only miR-24 following infection. The m169 transcript is cytoplasmic, capped, polyadenylated, and interacts with miRNA-27 through seed pairing: characteristic features of the normal messenger RNA (mRNA) targets of miRNAs. This virus-host interaction reveals a mode of miRNA regulation in which a mRNA directs the degradation of a miRNA. We speculate that RNA-mediated miRNA degradation could be a more general viral strategy for manipulating host cells.


Asunto(s)
MicroARNs/antagonistas & inhibidores , Muromegalovirus/genética , Regiones no Traducidas 3'/genética , Animales , Proteínas Argonautas/metabolismo , Secuencia de Bases , Sitios de Unión , Reactivos de Enlaces Cruzados/metabolismo , Citoplasma/metabolismo , ADN Complementario/genética , Regulación de la Expresión Génica/efectos de la radiación , Ensayos Analíticos de Alto Rendimiento , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Datos de Secuencia Molecular , Muromegalovirus/efectos de la radiación , Células 3T3 NIH , Nucleótidos/genética , Estabilidad del ARN/genética , Estabilidad del ARN/efectos de la radiación , Transporte de ARN/efectos de la radiación , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , Transcriptoma/genética , Rayos Ultravioleta
3.
Proc Natl Acad Sci U S A ; 107(31): 13830-5, 2010 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-20643939

RESUMEN

Although the functional parameters of microRNAs (miRNAs) have been explored in some depth, the roles of these molecules in viral infections remain elusive. Here we report a general method for global analysis of miRNA function that compares the significance of both overexpressing and inhibiting each mouse miRNA on the growth properties of different viruses. Our comparative analysis of representatives of all three herpesvirus subfamilies identified host miRNAs with broad anti- and proviral properties which extend to a single-stranded RNA virus. Specifically, we demonstrate the broad antiviral capacity of miR-199a-3p and illustrate that this individual host-encoded miRNA regulates multiple pathways required and/or activated by viruses, including PI3K/AKT and ERK/MAPK signaling, oxidative stress signaling, and prostaglandin synthesis. Global miRNA expression analysis further demonstrated that the miR-199a/miR-214 cluster is down-regulated in both murine and human cytomegalovirus infection and manifests similar antiviral properties in mouse and human cells. Overall, we report a general strategy for examining the contributions of individual host miRNAs in viral infection and provide evidence that these molecules confer broad inhibitory potential against multiple viruses.


Asunto(s)
Antivirales/análisis , Estudio de Asociación del Genoma Completo/métodos , Herpesviridae/efectos de los fármacos , MicroARNs/análisis , Animales , Antivirales/farmacología , Evaluación Preclínica de Medicamentos/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , MicroARNs/farmacología , Células 3T3 NIH , Transducción de Señal/efectos de los fármacos
4.
Vaccines (Basel) ; 7(4)2019 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-31717701

RESUMEN

Interferons (IFNs) play central roles in establishing innate immunity and mediating adaptive immunity against multiple pathogens. Three known types of IFNs identify their cognate receptors, initiate cascades of signalling events and eventually result in the induction of a myriad of IFN-stimulated genes (ISGs). These ISGs perform a multitude of functions and cumulatively corroborate a bespoke antiviral state to safeguard hosts against invading viruses. Owing to the unique nature of a chicken's immune system and the lack of foundational profiling information on the nature and dynamic expression of IFN-specific ISGs at the genome scale, we performed a systematic and extensive analysis of type I, II and III IFN-induced genes in chicken. Employing pan-IFN responsive chicken fibroblasts coupled with transcriptomics, we observed an over-representation of up-regulated ISGs compared to down-regulated ISGs by all types of IFNs. Intriguingly, prediction of IFN-stimulated response element (ISRE) and gamma-IFN activation sequence (GAS) revealed a substantial number of GAS motifs in selective and significantly induced ISGs in chicken. Extensive comparative, genome-wide and differential expression analysis of ISGs under equivalent signalling input catalogue a set of genes that were either IFN-specific or independent of types of IFNs used to prime fibroblasts. These comprehensive datasets, first of their kinds in chicken, will establish foundations to elucidate the mechanisms of actions and breadth of antiviral action of ISGs, which may propose alternative avenues for targeted antiviral therapy against viruses of poultry of public health importance.

5.
Genes (Basel) ; 10(2)2019 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-30769908

RESUMEN

Interferons (IFNs) are pleiotropic cytokines that establish a first line of defense against viral infections in vertebrates. Several types of IFN have been identified; however, limited information is available in poultry, especially using live animal experimental models. IFN-lambda (IFN-λ) has recently been shown to exert a significant antiviral impact against viral pathogens in mammals. In order to investigate the in vivo potential of chicken IFN-λ (chIFN-λ) as a regulator of innate immunity, and potential antiviral therapeutics, we profiled the transcriptome of chIFN-λ-stimulated chicken immune organs (in vivo) and compared it with primary chicken embryo fibroblasts (in vitro). Employing the baculovirus expression vector system (BEVS), recombinant chIFN-λ3 (rchIFN-λ3) was produced and its biological activities were demonstrated. The rchIFNλ3 induced a great array of IFN-regulated genes in primary chicken fibroblast cells. The transcriptional profiling using RNA-seq and subsequent bioinformatics analysis (gene ontology, differential expressed genes, and KEGGs analysis) of the bursa of Fabricious and the thymus demonstrated an upregulation of crucial immune genes (viperin, IKKB, CCL5, IL1ß, and AP1) as well as the antiviral signaling pathways. Interestingly, this experimental approach revealed contrasting evidence of the antiviral potential of chIFN-λ in both in vivo and in vitro models. Taken together, our data signifies the potential of chIFN-λ as a potent antiviral cytokine and highlights its future possible use as an antiviral therapeutic in poultry.


Asunto(s)
Pollos/inmunología , Citocinas/genética , Inmunidad Innata/genética , Interferones/genética , Animales , Antivirales/farmacología , Bolsa de Fabricio/inmunología , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Embrión de Pollo , Pollos/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Genes Reguladores/genética , Timo/inmunología , Replicación Viral/genética , Replicación Viral/inmunología
6.
Front Immunol ; 9: 2025, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30271403

RESUMEN

Innate antiviral immunity establishes first line of defense against invading pathogens through sensing their molecular structures such as viral RNA. This antiviral potential of innate immunity is mainly attributed to a myriad of IFN-stimulated genes (ISGs). Amongst well-characterized ISGs, we have previously shown that antiviral potential of chicken IFN-induced proteins with tetratricopeptides repeats 5 (chIFIT5) is determined by its interaction potential with 5'ppp containing viral RNA. Here, we generated transgenic chickens using avian sarcoma-leukosis virus (RCAS)-based gene transfer system that constitutively and stably express chIFIT5. The transgenic chickens infected with clinical dose (EID50 104 for HPAIV and 105 EID50 for vNDV) of high pathogenicity avian influenza virus (HPAIV; H5N1) or velogenic strain of Newcastle disease virus (vNDV; Genotype VII) showed marked resistance against infections. While transgenic chickens failed to sustain a lethal dose of these viruses (EID50 105 for HPAIV and 106 EID50 for vNDV), a delayed and lower level of clinical disease and mortality, reduced virus shedding and tissue damage was observed compared to non-transgenic control chickens. These observations suggest that stable expression of chIFIT5 alone is potentially insufficient in providing sterile protection against these highly virulent viruses; however, it is sufficient to ameliorate the clinical outcome of these RNA viruses. These findings propose the potential of innate immune genes in conferring genetic resistance in chickens against highly pathogenic and zoonotic viral pathogens causing sever disease in both animals and humans.


Asunto(s)
Proteínas Aviares/genética , Pollos/virología , Virus de la Influenza A/fisiología , Gripe Aviar/metabolismo , Factores Reguladores del Interferón/genética , Enfermedad de Newcastle/metabolismo , Virus de la Enfermedad de Newcastle/fisiología , Animales , Animales Modificados Genéticamente , Proteínas Aviares/metabolismo , Resistencia a la Enfermedad , Vectores Genéticos , Humanos , Inmunidad Innata , Gripe Aviar/genética , Gripe Humana/genética , Gripe Humana/metabolismo , Factores Reguladores del Interferón/metabolismo , Enfermedad de Newcastle/genética , Unión Proteica , ARN Viral/metabolismo , Virulencia , Zoonosis
7.
Sci Rep ; 8(1): 6794, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29717152

RESUMEN

The intracellular actions of interferon (IFN)-regulated proteins, including IFN-induced proteins with tetratricopeptide repeats (IFITs), attribute a major component of the protective antiviral host defense. Here we applied genomics approaches to annotate the chicken IFIT locus and currently identified a single IFIT (chIFIT5) gene. The profound transcriptional level of this effector of innate immunity was mapped within its unique cis-acting elements. This highly virus- and IFN-responsive chIFIT5 protein interacted with negative sense viral RNA structures that carried a triphosphate group on its 5' terminus (ppp-RNA). This interaction reduced the replication of RNA viruses in lentivirus-mediated IFIT5-stable chicken fibroblasts whereas CRISPR/Cas9-edited chIFIT5 gene knockout fibroblasts supported the replication of RNA viruses. Finally, we generated mosaic transgenic chicken embryos stably expressing chIFIT5 protein or knocked-down for endogenous chIFIT5 gene. Replication kinetics of RNA viruses in these transgenic chicken embryos demonstrated the antiviral potential of chIFIT5 in ovo. Taken together, these findings propose that IFIT5 specifically antagonize RNA viruses by sequestering viral nucleic acids in chickens, which are unique in innate immune sensing and responses to viruses of both poultry and human health significance.


Asunto(s)
Proteínas Aviares/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Factores Reguladores del Interferón/genética , Virus de la Enfermedad de Newcastle/inmunología , Vesiculovirus/inmunología , Secuencia de Aminoácidos , Animales , Proteínas Aviares/inmunología , Sistemas CRISPR-Cas , Pollos , Embrión no Mamífero , Fibroblastos/inmunología , Fibroblastos/virología , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Factores Reguladores del Interferón/inmunología , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/patogenicidad , Cultivo Primario de Células , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Repeticiones de Tetratricopéptidos , Transcripción Genética , Vesiculovirus/genética , Vesiculovirus/patogenicidad , Replicación Viral/inmunología
8.
Front Immunol ; 8: 49, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28197148

RESUMEN

Interferon (IFN) responses, mediated by a myriad of IFN-stimulated genes (ISGs), are the most profound innate immune responses against viruses. Cumulatively, these IFN effectors establish a multilayered antiviral state to safeguard the host against invading viral pathogens. Considerable genetic and functional characterizations of mammalian IFNs and their effectors have been made, and our understanding on the avian IFNs has started to expand. Similar to mammalian counterparts, three types of IFNs have been genetically characterized in most avian species with available annotated genomes. Intriguingly, chickens are capable of mounting potent innate immune responses upon various stimuli in the absence of essential components of IFN pathways including retinoic acid-inducible gene I, IFN regulatory factor 3 (IRF3), and possibility IRF9. Understanding these unique properties of the chicken IFN system would propose valuable targets for the development of potential therapeutics for a broader range of viruses of both veterinary and zoonotic importance. This review outlines recent developments in the roles of avian IFNs and ISGs against viruses and highlights important areas of research toward our understanding of the antiviral functions of IFN effectors against viral infections in birds.

9.
Sci Rep ; 7(1): 2719, 2017 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-28578423

RESUMEN

Interferons (IFNs) are essential components of the host innate immune system and define first-line of defence against pathogens. In mammals, several type I IFNs are identified, however, only limited data is available on the repertoire of IFNs in avian species. Here we report the characterization of chicken IFN-κ (chIFN-κ) near the type I IFN locus on the sex-determining Z chromosome. Genetic, evolutionary and syntenic analyses indicate that chIFN-κ is a type I IFN with conserved genetic features and promoter binding sites. chIFN-κ regulated the IFN-stimulated response element signalling pathways and activated a panel of IFN-regulated genes, antiviral mediators and transcriptional regulators. Priming of chicken primary fibroblasts and tracheal organ cultures with chIFN-κ imparted cellular protections against viral infections both in vitro and ex vivo. To determine whether chIFN-κ defines the antiviral state in developing chicken embryos, we used replication-competent retroviral RCAS vector system to generate transgenic chicken embryos that constitutively and stably expressed chIFN-κ. We could demonstrate that chIFN-κ markedly inhibited the replication of avian RNA viruses in ovo. Collectively, these results shed the light on the repertoire of IFNs in avian species and provide functional data on the interaction of the chIFN-κ with RNA viruses of poultry and public health importance.


Asunto(s)
Citocinas/genética , Interferón Tipo I/genética , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Antivirales/farmacología , Secuencia de Bases , Línea Celular , Embrión de Pollo , Pollos , Clonación Molecular , Citocinas/química , Citocinas/metabolismo , Citocinas/farmacología , Genómica/métodos , Interferón Tipo I/química , Interferón Tipo I/metabolismo , Interferón Tipo I/farmacología , Modelos Moleculares , Filogenia , Conformación Proteica , Virus ARN/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA