Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Methods Mol Biol ; 2824: 1-14, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39039402

RESUMEN

Rift Valley fever virus (RVFV) is a pathogen transmitted to humans and livestock via mosquito bites. This virus, which was discovered in Kenya in 1930, is considered by the World Health Organization (WHO) and the World Organisation for Animal Health (WOAH) to be associated with a high risk of causing large-scale epidemics. However, means dedicated to fighting RVFV have been limited, and despite recent research efforts, the virus remains poorly understood at both the molecular and cellular levels as well as at a broader scale of research in the field and in animal and human populations. In this introductory chapter of a methods book, we aim to provide readers with a concise overview of RVFV, from its ecology and transmission to the structural and genomic organization of virions and its life cycle in host cells.


Asunto(s)
Fiebre del Valle del Rift , Virus de la Fiebre del Valle del Rift , Virus de la Fiebre del Valle del Rift/genética , Fiebre del Valle del Rift/transmisión , Fiebre del Valle del Rift/virología , Animales , Humanos , Genoma Viral
2.
J Virol ; 86(20): 11333-44, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22896612

RESUMEN

Rift Valley fever virus (RVFV) is a highly pathogenic Phlebovirus that infects humans and ruminants. Initially confined to Africa, RVFV has spread outside Africa and presently represents a high risk to other geographic regions. It is responsible for high fatality rates in sheep and cattle. In humans, RVFV can induce hepatitis, encephalitis, retinitis, or fatal hemorrhagic fever. The nonstructural NSs protein that is the major virulence factor is found in the nuclei of infected cells where it associates with cellular transcription factors and cofactors. In previous work, we have shown that NSs interacts with the promoter region of the beta interferon gene abnormally maintaining the promoter in a repressed state. In this work, we performed a genome-wide analysis of the interactions between NSs and the host genome using a genome-wide chromatin immunoprecipitation combined with promoter sequence microarray, the ChIP-on-chip technique. Several cellular promoter regions were identified as significantly interacting with NSs, and the establishment of NSs interactions with these regions was often found linked to deregulation of expression of the corresponding genes. Among annotated NSs-interacting genes were present not only genes regulating innate immunity and inflammation but also genes regulating cellular pathways that have not yet been identified as targeted by RVFV. Several of these pathways, such as cell adhesion, axonal guidance, development, and coagulation were closely related to RVFV-induced disorders. In particular, we show in this work that NSs targeted and modified the expression of genes coding for coagulation factors, demonstrating for the first time that this hemorrhagic virus impairs the host coagulation cascade at the transcriptional level.


Asunto(s)
Factores de Coagulación Sanguínea/genética , ADN/genética , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos , Virus de la Fiebre del Valle del Rift/genética , Virus de la Fiebre del Valle del Rift/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Chlorocebus aethiops , Inmunoprecipitación de Cromatina , ADN/metabolismo , Estudio de Asociación del Genoma Completo , Interacciones Huésped-Patógeno/genética , Interferón beta/genética , Análisis por Matrices de Proteínas , ARN Mensajero/genética , Fiebre del Valle del Rift/genética , Fiebre del Valle del Rift/patología , Virus de la Fiebre del Valle del Rift/patogenicidad , Transcripción Genética , Células Vero , Proteínas no Estructurales Virales/análisis
4.
Emerg Infect Dis ; 18(6): 969-71, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22608405

RESUMEN

As further confirmation of a first human case of Rift Valley fever in 2007 in Comoros, we isolated Rift Valley fever virus in suspected human cases. These viruses are genetically closely linked to the 2006-2007 isolates from Kenya.


Asunto(s)
Genoma Viral , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/genética , Teorema de Bayes , Comoras , Humanos , Funciones de Verosimilitud , Datos de Secuencia Molecular , Filogenia , Virus de la Fiebre del Valle del Rift/aislamiento & purificación , Análisis de Secuencia de ADN
5.
J Gen Virol ; 93(Pt 7): 1456-1464, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22513390

RESUMEN

Currently, there are no worldwide licensed vaccines for Rift Valley fever (RVF) that are both safe and effective. Development and evaluation of vaccines, diagnostics and treatments depend on the availability of appropriate animal models. Animal models are also necessary to understand the basic pathobiology of infection. Here, we report the use of an inbred MBT/Pas mouse model that consistently reproduces RVF disease and serves our purpose for testing the efficacy of vaccine candidates; an attenuated Rift Valley fever virus (RVFV) and a recombinant RVFV-capripoxvirus. We show that this model is relevant for vaccine testing.


Asunto(s)
Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Fiebre del Valle del Rift/inmunología , Fiebre del Valle del Rift/prevención & control , Virus de la Fiebre del Valle del Rift/inmunología , Vacunación/métodos , Vacunas Virales/inmunología , Animales , Femenino , Humanos , Ratones , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas Virales/administración & dosificación
6.
J Virol ; 85(23): 12134-45, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21917943

RESUMEN

Rift Valley fever (RVF) is a mosquito-borne zoonotic disease caused by a phlebovirus of the family Bunyaviridae, which affects humans and ruminants in Africa and the Middle East. RFV virus (RVFV) possesses a single-stranded tripartite RNA genome of negative/ambisense polarity. The S segment utilizes the ambisense strategy and codes for two proteins, the N nucleoprotein and the nonstructural NSs protein, in opposite orientations. The two open reading frames (ORFs) are separated by an intergenic region (IGR) highly conserved among strains and containing a motif, 5'-GCUGC-3', present on the genome and antigenome, which was shown previously to play a role in transcription termination (C. G. Albarino, B. H. Bird, and S. T. Nichol, J. Virol. 81:5246-5256, 2007; T. Ikegami, S. Won, C. J. Peters, and S. Makino, J. Virol. 81:8421-8438, 2007). Here, we created recombinant RVFVs with mutations or deletions in the IGR and showed that the substitution of the motif sequence by a series of five A's inactivated transcription termination at the wild-type site but allowed the transcriptase to recognize another site with the consensus sequence present in the opposite ORF. Similar situations were observed for mutants in which the motif was still present in the IGR but located close to the stop codon of the translated ORF, supporting a model in which transcription is coupled to translation and translocating ribosomes abrogate transcription termination. Our data also showed that the signal tolerated some sequence variations, since mutation into 5'-GCAGC-3' was functional, and 5'-GUAGC-3' is likely the signal for the termination of the 3' end of the L mRNA.


Asunto(s)
ADN Intergénico/genética , Regulación Viral de la Expresión Génica , Genoma Viral , Virus de la Fiebre del Valle del Rift/genética , Regiones Terminadoras Genéticas/genética , Transcripción Genética , África , Animales , Secuencia de Bases , Northern Blotting , Chlorocebus aethiops , ADN Viral/genética , Datos de Secuencia Molecular , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Viral/genética , Fiebre del Valle del Rift/genética , Fiebre del Valle del Rift/virología , Homología de Secuencia de Ácido Nucleico , Células Vero
7.
J Virol ; 85(22): 12093-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21880754

RESUMEN

Lassa virus (LASV; Arenaviridae) is responsible for severe hemorrhagic fevers in Africa. LASV nucleoprotein (NP) plays important roles in regulating viral transcription and replication and in inhibiting type I interferon (IFN) production. The NP C-terminal domain contains a 3'-to-5' exonuclease activity involved in suppressing IFN induction. We have established a murine polymerase (Pol) I reverse genetics system for LASV, showing that residues D389 and G392 of NP were critical for LASV viability, while the D389A/G392A and D389T/392A double mutants were severely altered in the ability to suppress IFN in macrophages and dendritic cells. Assessing their attenuation in vivo may open new perspectives in vaccinology.


Asunto(s)
Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Células Dendríticas/inmunología , Interferón Tipo I/metabolismo , Virus Lassa/genética , Virus Lassa/inmunología , Macrófagos/inmunología , Sustitución de Aminoácidos , Animales , Chlorocebus aethiops , Células Dendríticas/virología , Virus Lassa/crecimiento & desarrollo , Macrófagos/virología , Datos de Secuencia Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/inmunología , Mutación Missense , Análisis de Secuencia de ADN , Células Vero , Carga Viral , Ensayo de Placa Viral
8.
J Immunol ; 185(10): 6146-56, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20937849

RESUMEN

Rift Valley fever (RVF) is an arthropod-borne viral disease repeatedly reported in many African countries and, more recently, in Saudi Arabia and Yemen. RVF virus (RVFV) primarily infects domesticated ruminants, resulting in miscarriage in pregnant females and death for newborns and young animals. It also has the ability to infect humans, causing a feverish syndrome, meningoencephalitis, or hemorrhagic fever. The various outcomes of RVFV infection in animals and humans argue for the existence of host genetic determinants controlling the disease. We investigated the susceptibility of inbred mouse strains to infection with the virulent RVFV ZH548 strain. Compared with classical BALB/cByJ mice, wild-derived Mus m. musculus MBT/Pas mice exhibited earlier and greater viremia and died sooner, a result in sharp contrast with their resistance to infection with West Nile virus and influenza A. Infection of mouse embryonic fibroblasts (MEFs) from MBT/Pas mice with RVFV also resulted in higher viral production. Microarray and quantitative RT-PCR experiments showed that BALB/cByJ MEFs displayed a significant activation of the type I IFN pathway. In contrast, MBT/Pas MEFs elicited a delayed and partial type I IFN response to RVFV infection. RNA interference-mediated inhibition of genes that were not induced by RVFV in MBT/Pas MEFs increased viral production in BALB/cByJ MEFs, thus demonstrating their functional importance in limiting viral replication. We conclude that the failure of MBT/Pas murine strain to induce, in due course, a complete innate immune response is instrumental in the selective susceptibility to RVF.


Asunto(s)
Inmunidad Innata/genética , Fiebre del Valle del Rift/genética , Fiebre del Valle del Rift/inmunología , Animales , Modelos Animales de Enfermedad , Fibroblastos/inmunología , Fibroblastos/virología , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Virol J ; 8: 249, 2011 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-21600011

RESUMEN

BACKGROUND: The genus Nairovirus in the family Bunyaviridae contains 34 tick-borne viruses classified into seven serogroups. Hazara virus (HAZV) belongs to the Crimean-Congo hemorrhagic fever (CCHF) serogroup that also includes CCHF virus (CCHFV) a major pathogen for humans. HAZV is an interesting model to study CCHFV due to a close serological and phylogenetical relationship and a classification which allows handling in a BSL2 laboratory. Nairoviruses are characterized by a tripartite negative-sense single stranded RNA genome (named L, M and S segments) that encode the RNA polymerase, the Gn-Gc glycoproteins and the nucleoprotein (NP), respectively. Currently, there are neither vaccines nor effective therapies for the treatment of any bunyavirus infection in humans. In this study we report, for the first time, the use of RNA interference (RNAi) as an approach to inhibit nairovirus replication. RESULTS: Chemically synthesized siRNAs were designed to target the mRNA produced by the three genomic segments. We first demonstrated that the siRNAs targeting the NP mRNA displayed a stronger antiviral effect than those complementary to the L and M transcripts in A549 cells. We further characterized the two most efficient siRNAs showing, that the induced inhibition is specific and associated with a decrease in NP synthesis during HAZV infection. Furthermore, both siRNAs depicted an antiviral activity when used before and after HAZV infection. We next showed that HAZV was sensitive to ribavirin which is also known to inhibit CCHFV. Finally, we demonstrated the additive or synergistic antiviral effect of siRNAs used in combination with ribavirin. CONCLUSIONS: Our study highlights the interest of using RNAi (alone or in combination with ribavirin) to treat nairovirus infection. This approach has to be considered for the development of future antiviral compounds targeting CCHFV, the most pathogenic nairovirus.


Asunto(s)
Antivirales/farmacología , Productos Biológicos/farmacología , Nairovirus/efectos de los fármacos , Nairovirus/fisiología , ARN Interferente Pequeño/farmacología , Ribavirina/farmacología , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Chlorocebus aethiops , Sinergismo Farmacológico , Humanos , Pruebas de Sensibilidad Microbiana
10.
J Gen Virol ; 91(Pt 1): 189-98, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19812268

RESUMEN

Crimean-Congo hemorrhagic fever virus (CCHFV) is a highly pathogenic, tick-borne member of the family Bunyaviridae and the genus Nairovirus. To better elucidate the pathogenesis of CCHFV, we analysed the host innate immune response induced in antigen-presenting cells (APCs) infected in vitro by CCHFV. Monocyte-derived dendritic cells (DCs) and macrophages (MPs) were both shown to be permissive for CCHFV and to replicate the virus, as monitored by genomic and antigenomic strand quantification. Virus replication was, however, controlled, corroborating an efficient alpha interferon-induced response. The upregulation of CD-83 and CD-86 indicated that CCHFV induced a partial maturation of DCs, which were also shown to activate the secretion of interleukin (IL)-6 and IL-8, but no tumour necrosis factor alpha (TNF-alpha). On the other hand, in MPs, CCHFV infection elicited a high IL-6 and TNF-alpha response and a moderate chemokine response. Nevertheless, when we compared these APC responses with those seen after infection with Dugbe virus (DUGV), a mildly pathogenic virus genetically close to CCHFV, we found that, in spite of some similarities, DUGV induced a higher cytokine/chemokine response in MPs. These results suggest that CCHFV is able to inhibit the activation of inflammatory mediators selectively in infection in vitro and that these differences could be relevant in pathogenesis.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/virología , Regulación de la Expresión Génica , Virus de la Fiebre Hemorrágica de Crimea-Congo/inmunología , Virus de la Fiebre Hemorrágica de Crimea-Congo/patogenicidad , Nairovirus/inmunología , Nairovirus/patogenicidad , Antígenos CD/biosíntesis , Antígeno B7-2/biosíntesis , Células Cultivadas , Quimiocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/virología , Humanos , Inmunoglobulinas/biosíntesis , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Macrófagos/inmunología , Macrófagos/virología , Glicoproteínas de Membrana/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Replicación Viral , Antígeno CD83
12.
J Virol ; 83(11): 5735-48, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19297476

RESUMEN

In their vertebrate hosts, arboviruses such as Semliki Forest virus (SFV) (Togaviridae) generally counteract innate defenses and trigger cell death. In contrast, in mosquito cells, following an early phase of efficient virus production, a persistent infection with low levels of virus production is established. Whether arboviruses counteract RNA interference (RNAi), which provides an important antiviral defense system in mosquitoes, is an important question. Here we show that in Aedes albopictus-derived mosquito cells, SFV cannot prevent the establishment of an antiviral RNAi response or prevent the spread of protective antiviral double-stranded RNA/small interfering RNA (siRNA) from cell to cell, which can inhibit the replication of incoming virus. The expression of tombusvirus siRNA-binding protein p19 by SFV strongly enhanced virus spread between cultured cells rather than virus replication in initially infected cells. Our results indicate that the spread of the RNAi signal contributes to limiting virus dissemination.


Asunto(s)
Culicidae/virología , Interferencia de ARN , Virus de los Bosques Semliki/genética , Animales , Línea Celular , Cricetinae , Regulación Viral de la Expresión Génica , ARN Viral/genética , Replicación Viral
13.
PLoS Pathog ; 4(1): e13, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18225953

RESUMEN

Rift Valley fever virus (RVFV) nonstructural protein NSs acts as the major determinant of virulence by antagonizing interferon beta (IFN-beta) gene expression. We demonstrate here that NSs interacts with the host protein SAP30, which belongs to Sin3A/NCoR/HDACs repressor complexes and interacts with the transcription factor YY1 that regulates IFN-beta gene expression. Using confocal microscopy and chromatin immunoprecipitation, we show that SAP30, YY1, and Sin3A-associated corepressor factors strongly colocalize with nuclear NSs filaments and that NSs, SAP30 and Sin3A-associated factors are recruited on the IFN-beta promoter through YY1, inhibiting CBP recruitment, histone acetylation, and transcriptional activation. To ascertain the role of SAP30, we produced, by reverse genetics, a recombinant RVFV in which the interacting domain in NSs was deleted. The virus was unable to inhibit the IFN response and was avirulent for mice. We discuss here the strategy developed by the highly pathogenic RVFV to evade the host antiviral response, affecting nuclear organization and IFN-beta promoter chromatin structure.


Asunto(s)
Histona Desacetilasas/metabolismo , Interferón beta/metabolismo , Proteínas Represoras/metabolismo , Virus de la Fiebre del Valle del Rift/fisiología , Proteínas no Estructurales Virales/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Núcleo Celular/metabolismo , Células Cultivadas , Chlorocebus aethiops , Regulación Viral de la Expresión Génica , Histona Desacetilasas/genética , Interferón beta/genética , Ratones , Microscopía Confocal , Mutación , Complejo Correpresor Histona Desacetilasa y Sin3 , Técnicas del Sistema de Dos Híbridos , Células Vero , Proteínas no Estructurales Virales/genética , Virulencia
14.
Nucleic Acids Res ; 36(12): e72, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18515351

RESUMEN

DNA complementarity is expressed by way of three hydrogen bonds for a G:C base pair and two for A:T. As a result, careful control of the denaturation temperature of PCR allows selective amplification of AT-rich alleles. Yet for the same reason, the converse is not possible, selective amplification of GC-rich alleles. Inosine (I) hydrogen bonds to cytosine by two hydrogen bonds while diaminopurine (D) forms three hydrogen bonds with thymine. By substituting dATP by dDTP and dGTP by dITP in a PCR reaction, DNA is obtained in which the natural hydrogen bonding rule is inversed. When PCR is performed at limiting denaturation temperatures, it is possible to recover GC-rich viral genomes and inverted Alu elements embedded in cellular mRNAs resulting from editing by dsRNA dependent host cell adenosine deaminases. The editing of Alu elements in cellular mRNAs was strongly enhanced by type I interferon induction indicating a novel link mRNA metabolism and innate immunity.


Asunto(s)
Adenosina Desaminasa/metabolismo , ADN/química , Secuencia Rica en GC , Reacción en Cadena de la Polimerasa/métodos , Edición de ARN , 2-Aminopurina/análogos & derivados , 2-Aminopurina/química , Secuencia Rica en At , Elementos Alu , Animales , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Reordenamiento Génico de Linfocito B , Enlace de Hidrógeno , Región Variable de Inmunoglobulina/genética , Inosina Trifosfato/química , Virus del Sarampión/genética , Datos de Secuencia Molecular , Desnaturalización de Ácido Nucleico , ARN Mensajero/química , Proteínas de Unión al ARN , Células Vero
15.
Nat Commun ; 11(1): 3281, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32612175

RESUMEN

Amyloid fibrils result from the aggregation of host cell-encoded proteins, many giving rise to specific human illnesses such as Alzheimer's disease. Here we show that the major virulence factor of Rift Valley fever virus, the protein NSs, forms filamentous structures in the brain of mice and affects mortality. NSs assembles into nuclear and cytosolic disulfide bond-dependent fibrillary aggregates in infected cells. NSs structural arrangements exhibit characteristics typical for amyloids, such as an ultrastructure of 12 nm-width fibrils, a strong detergent resistance, and interactions with the amyloid-binding dye Thioflavin-S. The assembly dynamics of viral amyloid-like fibrils can be visualized in real-time. They form spontaneously and grow in an amyloid fashion within 5 hours. Together, our results demonstrate that viruses can encode amyloid-like fibril-forming proteins and have strong implications for future research on amyloid aggregation and toxicity in general.


Asunto(s)
Amiloide/metabolismo , Proteínas Amiloidogénicas/metabolismo , Fiebre del Valle del Rift/metabolismo , Virus de la Fiebre del Valle del Rift/metabolismo , Proteínas no Estructurales Virales/metabolismo , Amiloide/química , Amiloide/ultraestructura , Proteínas Amiloidogénicas/química , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Núcleo Celular/virología , Chlorocebus aethiops , Células HeLa , Humanos , Ratones , Microscopía Confocal , Microscopía Electrónica de Transmisión , Agregación Patológica de Proteínas/metabolismo , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/patogenicidad , Células Vero , Proteínas no Estructurales Virales/química , Virulencia , Factores de Virulencia
16.
Virol J ; 6: 6, 2009 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-19149901

RESUMEN

BACKGROUND: Affecting both livestock and humans, Rift Valley Fever is considered as one of the most important viral zoonoses in Africa. However, no licensed vaccines or effective treatments are yet available for human use. Naked DNA vaccines are an interesting approach since the virus is highly infectious and existing attenuated Rift Valley Fever virus vaccine strains display adverse effects in animal trials. In this study, gene-gun immunisations with cDNA encoding structural proteins of the Rift Valley Fever virus were evaluated in mice. The induced immune responses were analysed for the ability to protect mice against virus challenge. RESULTS: Immunisation with cDNA encoding the nucleocapsid protein induced strong humoral and lymphocyte proliferative immune responses, and virus neutralising antibodies were acquired after vaccination with cDNA encoding the glycoproteins. Even though complete protection was not achieved by genetic immunisation, four out of eight, and five out of eight mice vaccinated with cDNA encoding the nucleocapsid protein or the glycoproteins, respectively, displayed no clinical signs of infection after challenge. In contrast, all fourteen control animals displayed clinical manifestations of Rift Valley Fever after challenge. CONCLUSION: The appearance of Rift Valley Fever associated clinical signs were significantly decreased among the DNA vaccinated mice and further adjustment of this strategy may result in full protection against Rift Valley Fever.


Asunto(s)
Fiebre del Valle del Rift/inmunología , Fiebre del Valle del Rift/prevención & control , Virus de la Fiebre del Valle del Rift/inmunología , Vacunas de ADN/administración & dosificación , Vacunas Virales/administración & dosificación , Animales , Anticuerpos Antivirales/sangre , Biolística , Línea Celular , ADN Complementario/genética , ADN Complementario/inmunología , Femenino , Glicoproteínas/genética , Glicoproteínas/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas de la Nucleocápside/genética , Proteínas de la Nucleocápside/inmunología , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/genética , Vacunación , Vacunas de ADN/inmunología , Vacunas Virales/inmunología
17.
J Clin Microbiol ; 46(11): 3653-9, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18799705

RESUMEN

The development and validation of a one-step, single-tube, real-time accelerated reverse-transcription loop-mediated isothermal amplification (RT-LAMP) for the detection of the L RNA segment of Rift Valley fever virus (RVFV) are described. The assay was performed at a constant temperature (63 degrees C), with a real-time follow-up using a LightCycler and a double-stranded-DNA-intercalating fluorochrome. The assay is highly sensitive and comparable to real-time RT-PCR, with a detection limit of approximately 10 RNA copies per assay. However, the RT-LAMP assay is much faster than traditional RT-PCR and generates results in <30 min for most diluted samples. The specificity of the primers was established using other, related arboviruses as well as virus-containing and virus-free sera. The RT-LAMP assay reported here is thus a valuable tool for the rapid detection of RVFV in field diagnostic laboratories.


Asunto(s)
Técnicas de Amplificación de Ácido Nucleico/métodos , Fiebre del Valle del Rift/diagnóstico , Virus de la Fiebre del Valle del Rift/aislamiento & purificación , Cartilla de ADN/genética , Humanos , ARN Viral/genética , Fiebre del Valle del Rift/virología , Sensibilidad y Especificidad , Factores de Tiempo
18.
Am J Trop Med Hyg ; 76(5): 827-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17488899

RESUMEN

Transmission experiments are essential to assess vector competence. In an attempt to study Rift Valley fever virus (RVFV) transmission in its arthropod vector, disseminated infection rates were measured in Culex pipiens quinquefasciatus exposed to Clone 13, an avirulent variant of RVFV. We compared the classic system of glass feeders covered with different membranes (0-1%) with an alternative system in which a cotton stick is used as a support of a blood meal. The latter system showed the highest successful feeding rate (80%). Using this system, we optimized two parameters to obtain the highest numbers of engorged females and thus the highest numbers of females with disseminated infection: 1) use of freshly collected washed erythrocytes and 2) duration of the extrinsic incubation period of at least 14 days after infection.


Asunto(s)
Culex/virología , Métodos de Alimentación/veterinaria , Insectos Vectores/virología , Virus de la Fiebre del Valle del Rift/patogenicidad , Animales , Eritrocitos , Métodos de Alimentación/instrumentación , Femenino , Fiebre del Valle del Rift/transmisión , Virus de la Fiebre del Valle del Rift/aislamiento & purificación , Ovinos , Factores de Tiempo
20.
Curr Mol Med ; 5(8): 827-34, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16375716

RESUMEN

Rift Valley fever is considered to be one of the most important viral zoonoses in Africa. In 2000, the Rift valley fever virus spread to the Arabian Peninsula and caused two simultaneous outbreaks in Yemen and Saudi Arabia. It is transmitted to ruminants and to humans by mosquitoes. The viral agent is an arbovirus, which belongs to the Phlebovirus genus in the Bunyaviridae family. This family of viruses comprises more than 300 members grouped into five genera: Orthobunyavirus, Phlebovirus, Hantavirus, Nairovirus, and Tospovirus. Several members of the Bunyaviridae family are responsible for fatal hemorrhagic fevers: Rift Valley fever virus (Phlebovirus), Crimean-Congo hemorrhagic fever virus (Nairovirus), Hantaan, Sin Nombre and related viruses (Hantavirus), and recently Garissa, now identified as Ngari virus (Orthobunyavirus). Here are reviewed recent advances in Rift Valley fever virus, its epidemiology, molecular biology and focus on recent data on the interactions between viral and cellular proteins, which help to understand the molecular mechanisms utilized by the virus to circumvent the host cellular response.


Asunto(s)
Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/patogenicidad , Animales , Vectores de Enfermedades , Humanos , Inmunidad , Salud Pública , Fiebre del Valle del Rift/diagnóstico , Fiebre del Valle del Rift/terapia , Virus de la Fiebre del Valle del Rift/clasificación , Virus de la Fiebre del Valle del Rift/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA