Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 7(11): e1002335, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22102808

RESUMEN

In nature, arthropod-borne viruses (arboviruses) perpetuate through alternating replication in vertebrate and invertebrate hosts. The trade-off hypothesis proposes that these viruses maintain adequate replicative fitness in two disparate hosts in exchange for superior fitness in one host. Releasing the virus from the constraints of a two-host cycle should thus facilitate adaptation to a single host. This theory has been addressed in a variety of systems, but remains poorly understood. We sought to determine the fitness implications of alternating host replication for West Nile virus (WNV) using an in vivo model system. Previously, WNV was serially or alternately passed 20 times in vivo in chicks or mosquitoes and resulting viruses were characterized genetically. In this study, these test viruses were competed in vivo in fitness assays against an unpassed marked reference virus. Fitness was assayed in chicks and in two important WNV vectors, Culex pipiens and Culex quinquefasciatus. Chick-specialized virus displayed clear fitness gains in chicks and in Cx. pipiens but not in Cx. quinquefasciatus. Cx. pipiens-specialized virus experienced reduced fitness in chicks and little change in either mosquito species. These data suggest that when fitness is measured in birds the trade-off hypothesis is supported; but in mosquitoes it is not. Overall, these results suggest that WNV evolution is driven by alternate cycles of genetic expansion in mosquitoes, where purifying selection is weak and genetic diversity generated, and restriction in birds, where purifying selection is strong.


Asunto(s)
Aptitud Genética , Variación Genética , Interacciones Huésped-Patógeno , Virus del Nilo Occidental/genética , Virus del Nilo Occidental/fisiología , Animales , Evolución Biológica , Pollos/virología , Culex/virología , Interacciones Huésped-Patógeno/genética , Selección Genética , Pase Seriado , Fiebre del Nilo Occidental/virología
2.
J Virol ; 85(23): 12605-13, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21937657

RESUMEN

West Nile virus (WNV) is similar to other RNA viruses in that it forms genetically complex populations within hosts. The virus is maintained in nature in mosquitoes and birds, with each host type exerting distinct influences on virus populations. We previously observed that prolonged replication in mosquitoes led to increases in WNV genetic diversity and diminished pathogenesis in mice without remarkable changes to the consensus genome sequence. We therefore sought to evaluate the relationships between individual and group phenotypes in WNV and to discover novel viral determinants of pathogenesis in mice and fitness in mosquitoes and birds. Individual plaque size variants were isolated from a genetically complex population, and mutations conferring a small-plaque and mouse-attenuated phenotype were localized to the RNA helicase domain of the NS3 protein by reverse genetics. The mutation, an Asp deletion, did not alter type I interferon production in the host but rendered mutant viruses more susceptible to interferon compared to wild type (WT) WNV. Finally, we used an in vivo fitness assay in Culex quinquefasciatus mosquitoes and chickens to determine whether the mutation in NS3 influenced fitness. The fitness of the NS3 mutant was dramatically lower in chickens and moderately lower in mosquitoes, indicating that RNA helicase is a major fitness determinant of WNV and that the effect on fitness is host specific. Overall, this work highlights the complex relationships that exist between individual and group phenotypes in RNA viruses and identifies RNA helicase as an attenuation and fitness determinant in WNV.


Asunto(s)
Pollos/virología , Culicidae/virología , Genoma Viral , Fiebre del Nilo Occidental/patología , Fiebre del Nilo Occidental/parasitología , Virus del Nilo Occidental/genética , Virus del Nilo Occidental/patogenicidad , Animales , Células Cultivadas , Pollos/genética , Chlorocebus aethiops , Cricetinae , Culicidae/genética , Culicidae/patogenicidad , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/virología , Variación Genética , Interferones/metabolismo , Riñón/citología , Riñón/metabolismo , Riñón/virología , Ratones , Ratones Endogámicos C3H , Mutación/genética , ARN Helicasas/genética , ARN Helicasas/metabolismo , ARN Viral/genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Células Vero , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Fiebre del Nilo Occidental/virología
3.
J Virol Methods ; 173(2): 378-83, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21295611

RESUMEN

Reverse genetics approaches that enable the generation of recombinant influenza A viruses entirely from plasmids are invaluable for studies on virus replication, morphogenesis, pathogenesis, or transmission. Furthermore, influenza virus reverse genetics is now critical for the development of new vaccines for this human and animal pathogen. Periodically, influenza gene segments are unstable within plasmids in bacteria. The PB2 gene segment of a highly pathogenic avian H5 influenza virus A/Turkey/Ontario/7732/1966 (Ty/Ont) was unstable in commonly available cloning plasmids (e.g., pcDNA3.1/V5-His-TOPO) and in standard influenza virus reverse genetics plasmids (e.g., pHH21), which contain high copy origins of replication. Thus, a low-copy influenza reverse genetics plasmid (pGJ3C3) was developed to enable rapid cloning of unstable influenza A virus genes using ligation-independent recombination-based cloning. The unstable Ty/Ont PB2 gene segment was efficiently cloned using the pGJ3C3 plasmid and this clone was used to rescue a recombinant Ty/Ont virus. This low copy reverse genetics plasmid will be useful for cloning other unstable segments of influenza A viruses in order to rescue recombinant viruses, which will facilitate basic studies and vaccine seed stock production.


Asunto(s)
Ingeniería Genética/métodos , Genética Microbiana/métodos , Virus de la Influenza A/genética , Biología Molecular/métodos , Virología/métodos , Animales , Clonación Molecular/métodos , Dosificación de Gen , Vectores Genéticos , Inestabilidad Genómica , Humanos , Virus de la Influenza A/crecimiento & desarrollo , Plásmidos
4.
Infect Genet Evol ; 11(2): 460-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21215334

RESUMEN

St. Louis encephalitis virus (SLEV; Flaviviridae; Flavivirus) is a member of the Japanese encephalitis serocomplex and a close relative of West Nile virus (WNV). Although SLEV remains endemic to the US, both levels of activity and geographical dispersal are relatively constrained when compared to the widespread distribution of WNV. In recent years, WNV appears to have displaced SLEV in California, yet both viruses currently coexist in Texas and several other states. It has become clear that viral swarm characterization is required if we are to fully evaluate the relationship between viral genomes, viral evolution, and epidemiology. Mutant swarm size and composition may be particularly important for arboviruses, which require replication not only in diverse tissues but also divergent hosts. In order to evaluate temporal, spatial, and host-specific patterns in the SLEV mutant swarm, we determined the size, composition, and phylogeny of the intrahost swarm within primary mosquito isolates from both Texas and California. Results indicate a general trend of decreasing intrahost diversity over time in both locations, with recent isolates being highly genetically homogeneous. Additionally, phylogenic analyses provide detailed information on the relatedness of minority variants both within and among strains and demonstrate how both geographic isolation and seasonal maintenance have shaped the viral swarm. Overall, these data generally provide insight into how time, space, and unique transmission cycles influence the SLEV mutant swarm and how understanding these processes can ultimately lead to a better understanding of arbovirus evolution and epidemiology.


Asunto(s)
Culex/virología , Virus de la Encefalitis de San Luis/genética , Virus de la Encefalitis de San Luis/aislamiento & purificación , Especificidad del Huésped , Insectos Vectores/virología , Mutación , Animales , Aves/virología , California/epidemiología , Encefalitis de San Luis/epidemiología , Encefalitis de San Luis/transmisión , Encefalitis de San Luis/virología , Variación Genética , Genoma Viral , Humanos , Filogenia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia , Texas/epidemiología
5.
PLoS One ; 4(11): e7876, 2009 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-19924238

RESUMEN

St. Louis encephalitis virus (SLEV; Flaviviridae, flavivirus) was the major cause of epidemic flaviviral encephalitis in the U.S. prior to the introduction of West Nile virus (WNV) in 1999. However, outbreaks of SLEV have been significantly more limited then WNV in terms of levels of activity and geographic dispersal. One possible explanation for these variable levels of activity is that differences in the potential for each virus to adapt to its host cycle exist. The need for arboviruses to replicate in disparate hosts is thought to result in constraints on both evolution and host-specific adaptation. If cycling is the cause of genetic stability observed in nature and arboviruses lack host specialization, then sequential passage should result in both the accumulation of mutations and specialized viruses better suited for replication in that host. Previous studies suggest that WNV and SLEV differ in capacity for both genetic change and host specialization, and in the costs each accrues from specializing. In an attempt to clarify how selective pressures contribute to epidemiological patterns of WNV and SLEV, we evaluated mutant spectra size, consensus genetic change, and phenotypic changes for SLEV in vivo following 20 sequential passages via inoculation in either Culex pipiens mosquitoes or chickens. Results demonstrate that the capacity for genetic change is large for SLEV and that the size of the mutant spectrum is host-dependent using our passage methodology. Despite this, a general lack of consensus change resulted from passage in either host, a result that contrasts with the idea that constraints on evolution in nature result from host cycling alone. Results also suggest that a high level of adaptation to both hosts already exists, despite host cycling. A strain significantly more infectious in chickens did emerge from one lineage of chicken passage, yet other lineages and all mosquito passage strains did not display measurable host-specific fitness gains. In addition, increased infectivity in chickens did not decrease infectivity in mosquitoes, which further contrasts the concept of fitness trade-offs for arboviruses.


Asunto(s)
Virus de la Encefalitis de San Luis/metabolismo , Encefalitis de San Luis/transmisión , Animales , Evolución Biológica , Pollos , Chlorocebus aethiops , Clonación Molecular , Culicidae , Encefalitis de San Luis/veterinaria , Femenino , Variación Genética , Concentración 50 Inhibidora , Cinética , Análisis de Secuencia de ADN , Especificidad de la Especie , Células Vero
6.
Virology ; 374(2): 256-60, 2008 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-18395240

RESUMEN

To investigate differential evolutionary rates and selective forces of WNV in hosts and vectors, we measured the genetic diversity that arose during alternating passage in mosquitoes and birds. Within-host genetic diversity was monitored in each of three experimentally passed replicates, and the complete genome sequence of each WNV strain was determined after passage. The intrahost genetic diversity that arose during alternating passage was significantly greater than the diversity generated during chicken-only passage and similar to mosquito-only passage. dN/dS ratios suggested purifying selection similar to chick-passed virus, but not to mosquito-passed virus. Thus, the abundant genetic variation contributed to WNV populations through infection of mosquitoes and the strong purifying selection contributed by infection of birds may be maintained despite frequent host switching.


Asunto(s)
Variación Genética , Interacciones Huésped-Patógeno , Selección Genética , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/clasificación , Virus del Nilo Occidental/patogenicidad , Animales , Línea Celular , Pollos/virología , Cricetinae , Culex/virología , Femenino , Genoma Viral , Filogenia , Enfermedades de las Aves de Corral/virología , Análisis de Secuencia de ADN , Pase Seriado , Especificidad de la Especie , Organismos Libres de Patógenos Específicos , Fiebre del Nilo Occidental/veterinaria , Virus del Nilo Occidental/genética
7.
Virology ; 367(2): 339-47, 2007 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-17617432

RESUMEN

A small-plaque variant (SP) of West Nile virus (WNV) was isolated in Vero cell culture from kidney tissue of an American crow collected in New York in 2000. The in vitro growth of the SP and parental (WT) strains was characterized in mammalian (Vero), avian (DF-1 and PDE), and mosquito (C6/36) cells. The SP variant replicated less efficiently than did the WT in Vero cells. In avian cells, SP growth was severely restricted at high temperatures, suggesting that the variant is temperature sensitive. In mosquito cells, growth of SP and WT was similar, but in vivo in Culex pipiens (L.) there were substantial differences. Relative to WT, SP exhibited reduced replication following intrathoracic inoculation and lower infection, dissemination, and transmission rates following oral infection. Analysis of the full length sequence of the SP variant identified sequence differences which led to only two amino acid substitutions relative to WT, prM P54S and NS2A V61A.


Asunto(s)
Variación Genética , Replicación Viral/fisiología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/genética , Aedes/virología , Animales , Enfermedades de las Aves/virología , Chlorocebus aethiops , Cuervos/virología , Insectos Vectores/fisiología , Insectos Vectores/virología , New York/epidemiología , Temperatura , Células Vero , Fiebre del Nilo Occidental/epidemiología , Virus del Nilo Occidental/clasificación , Virus del Nilo Occidental/fisiología
8.
Virology ; 360(2): 469-76, 2007 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-17134731

RESUMEN

To define the impact of mosquitoes and birds on intrahost WNV population dynamics, the mutant spectra that arose as a result of 20 serial in vivo passages in Culex pipiens and young chickens were examined. Genetically homogeneous WNV was serially passaged 20 times in each host. Genetic diversity was greater in mosquito-passaged WNV compared to chicken-passaged WNV. Changes in the viral consensus sequence occurred in WNV passaged in mosquitoes earlier and more frequently than in chicken-passaged WNV. Analysis of synonymous and nonsynonymous variation suggested that purifying selection was relaxed during passage in mosquitoes. Mortality in mice was significantly negatively correlated with the size of the WNV mutant spectrum. These studies suggest that mosquitoes serve as sources for WNV genetic diversity, that birds are selective sieves, and that both the consensus sequence and the mutant spectrum contribute to WNV phenotype.


Asunto(s)
Variación Genética , Mutación , Fiebre del Nilo Occidental/mortalidad , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/genética , Virus del Nilo Occidental/patogenicidad , Animales , Pollos/virología , Culex/virología , Modelos Animales de Enfermedad , Femenino , Genoma Viral , Ratones , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Fiebre del Nilo Occidental/patología , Virus del Nilo Occidental/aislamiento & purificación
9.
Appl Environ Microbiol ; 72(4): 2460-70, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16597944

RESUMEN

We have developed sediment-free anaerobic enrichment cultures that dechlorinate a broad spectrum of highly chlorinated polychlorinated biphenyls (PCBs). The cultures were developed from Aroclor 1260-contaminated sediment from the Housatonic River in Lenox, MA. Sediment slurries were primed with 2,6-dibromobiphenyl to stimulate Process N dechlorination (primarily meta dechlorination), and sediment was gradually removed by successive transfers (10%) to minimal medium. The cultures grow on pyruvate, butyrate, or acetate plus H(2). Gas chromatography-electron capture detector analysis demonstrated that the cultures extensively dechlorinate 50 to 500 mug/ml of Aroclor 1260 at 22 to 24 degrees C by Dechlorination Process N. Triplicate cultures of the eighth transfer without sediment dechlorinated 76% of the hexa- through nonachlorobiphenyls in Aroclor 1260 (250 mug/ml) to tri- through pentachlorobiphenyls in 110 days. At least 64 PCB congeners, all of which are chlorinated on both rings and 47 of which have six or more chlorines, were substrates for this dechlorination. To characterize the bacterial diversity in the enrichments, we used eubacterial primers to amplify and clone 16S rRNA genes from DNA extracted from cultures grown on acetate plus H(2). Restriction fragment length polymorphism analysis of 107 clones demonstrated the presence of Thauera-like Betaproteobacteria, Geobacter-like Deltaproteobacteria, Pseudomonas species, various Clostridiales, Bacteroidetes, Dehalococcoides of the Chloroflexi group, and unclassified Eubacteria. Our development of highly enriched, robust, stable, sediment-free cultures that extensively dechlorinate a highly chlorinated commercial PCB mixture is a major and unprecedented breakthrough in the field. It will enable intensive study of the organisms and genes responsible for a major PCB dechlorination process that occurs in the environment and could also lead to effective remediation applications.


Asunto(s)
Arocloros/metabolismo , Bacterias Anaerobias/clasificación , Bacterias Anaerobias/crecimiento & desarrollo , Cloro/metabolismo , Contaminantes Ambientales/metabolismo , Arocloros/química , Bacterias Anaerobias/genética , Bacterias Anaerobias/metabolismo , Técnicas Bacteriológicas , Medios de Cultivo/química , ADN Bacteriano/análisis , ADN Ribosómico/análisis , Genes de ARNr , Sedimentos Geológicos , Datos de Secuencia Molecular , Bifenilos Policlorados/metabolismo , Polimorfismo de Longitud del Fragmento de Restricción , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN , Contaminantes del Suelo/metabolismo
10.
J Gen Virol ; 86(Pt 8): 2175-2183, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16033965

RESUMEN

Intrahost genetic diversity was analysed in naturally infected mosquitoes and birds to determine whether West Nile virus (WNV) exists in nature as a quasispecies and to quantify selective pressures within and between hosts. WNV was sampled from ten infected birds and ten infected mosquito pools collected on Long Island, NY, USA, during the peak of the 2003 WNV transmission season. A 1938 nt fragment comprising the 3' 1159 nt of the WNV envelope (E) coding region and the 5' 779 nt of the non-structural protein 1 (NS1) coding region was amplified and cloned and 20 clones per specimen were sequenced. Results from this analysis demonstrate that WNV infections are derived from a genetically diverse population of genomes in nature. The mean nucleotide diversity was 0.016 % within individual specimens and the mean percentage of clones that differed from the consensus sequence was 19.5 %. WNV sequences in mosquitoes were significantly more genetically diverse than WNV in birds. No host-dependent bias for particular types of mutations was observed and estimates of genetic diversity did not differ significantly between E and NS1 coding sequences. Non-consensus clones obtained from two avian specimens had highly similar genetic signatures, providing preliminary evidence that WNV genetic diversity may be maintained throughout the enzootic transmission cycle, rather than arising independently during each infection. Evidence of purifying selection was obtained from both intra- and interhost WNV populations. Combined, these data support the observation that WNV populations may be structured as a quasispecies and document strong purifying natural selection in WNV populations.


Asunto(s)
Aves/virología , Culicidae/virología , Variación Genética , ARN Viral , Virus del Nilo Occidental/genética , Animales , Datos de Secuencia Molecular , New York , Selección Genética , Especificidad de la Especie , Proteínas del Envoltorio Viral/genética , Proteínas no Estructurales Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA