Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Virol ; 97(7): e0065923, 2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37367225

RESUMEN

The specific packaging of the viral RNA genome into virus particles is an essential step in the replication cycle of coronaviruses (CoVs). Using a single-cycle, replicable severe acute respiratory syndrome CoV-2 (SARS-CoV-2) mutant, we demonstrated the preferential packaging of the SARS-CoV-2 genomic RNA into purified virus particles. Furthermore, based on the sequence of an efficiently packaged defective interfering RNA of SARS-CoV, a closely related CoV, that was generated after serial passages of SARS-CoV in cell culture, we designed a series of replication-competent SARS-CoV-2 minigenome RNAs to identify the specific viral RNA region that is important for SARS-CoV-2 RNA packaging into virus particles. We showed that a 1.4-kb-long sequence, derived from the nsp12 and nsp13 coding regions of the SARS-CoV-2 genomic RNA, is required for the efficient packaging of SARS-CoV-2 minigenome RNA into SARS-CoV-2 particles. In addition, we also showed that the presence of possibly the entire 1.4-kb-long sequence is important for the efficient packaging of SARS-CoV-2 RNA. Our findings highlight the differences between the RNA packaging sequence identified in SARS-CoV-2, a Sarbecovirus, and the packaging signal of mouse hepatitis virus (MHV), an Embecovirus, which is a 95-nt-long sequence located at the nsp15 coding region of MHV genomic RNA. Collectively, our data imply that both the location and the sequence/structural features of the RNA element(s) that drives the selective and efficient packaging of viral genomic RNA are not conserved among the subgenera Embecovirus and Sarbecovirus within the Betacoronavirus genus. IMPORTANCE Elucidating the mechanism of SARS-CoV-2 RNA packaging into virus particles is important for the rational design of antiviral drugs that inhibit this vital step in the replication cycle of CoVs. However, our knowledge about the RNA packaging mechanism in SARS-CoV-2, including the identification of the viral RNA region important for SARS-CoV-2 RNA packaging, is limited, primarily due to the logistical challenges of handing SARS-CoV-2 in biosafety level 3 (BSL3) facilities. Our study, using a single-cycle, replicable SARS-CoV-2 mutant, which can be handled in a BSL2 lab, demonstrated the preferential packaging of full-length SARS-CoV-2 genomic RNA into virus particles and identified a specific 1.4-kb-long RNA region in SARS-CoV-2 genomic RNA that is required for the efficient packaging of SARS-CoV-2 RNA into virus particles. The information generated in our study could be valuable for clarifying the mechanisms of SARS-CoV-2 RNA packaging and for the development of targeted therapeutics against SARS-CoV-2 and other related CoVs.


Asunto(s)
ARN Viral , SARS-CoV-2 , Empaquetamiento del Genoma Viral , Proteínas Virales , COVID-19/virología , Virus de la Hepatitis Murina/genética , Virus de la Hepatitis Murina/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Empaquetamiento del Genoma Viral/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
2.
J Virol ; 95(14): e0042921, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-33952635

RESUMEN

Rift Valley fever phlebovirus (RVFV) has a single-stranded, negative-sense RNA genome, consisting of L, M, and S segments. The virion carries two envelope glycoproteins, Gn and Gc, along with ribonucleoprotein complexes (RNPs), composed of encapsidated genomes carrying N protein and the viral polymerase, L protein. A quantitative analysis of the profile of viral RNA segments packaged into RVFV particles showed that all three genomic RNA segments had similar packaging abilities, whereas among antigenomic RNA segments, the antigenomic S RNA, which serves as the template for the transcription of mRNA expressing the RVFV virulence factor, NSs, displayed a significantly higher packaging ability. To delineate the factor(s) governing the packaging of RVFV RNA segments, we characterized the interactions between Gn and viral RNPs in RVFV-infected cells. Coimmunoprecipitation analysis demonstrated the interaction of Gn with N protein, L protein, and viral RNAs in RVFV-infected cells. Furthermore, UV-cross-linking and immunoprecipitation analysis revealed, for the first time in bunyaviruses, the presence of a direct interaction between Gn and all the viral RNA segments in RVFV-infected cells. Notably, analysis of the ability of Gn to bind to RVFV RNA segments indicated a positive correlation with their respective packaging abilities and highlighted a binding preference of Gn for antigenomic S RNA, among the antigenomic RNA segments, suggesting the presence of a selection mechanism for antigenomic S RNA incorporation into infectious RVFV particles. Collectively, the results of our study illuminate the importance of a direct interaction between Gn and viral RNA segments in determining their efficiency of incorporation into RVFV particles. IMPORTANCE Rift Valley fever phlebovirus, a bunyavirus, is a mosquito-borne, segmented RNA virus that can cause severe disease in humans and ruminants. An essential step in RVFV life cycle is the packaging of viral RNA segments to produce infectious virus particles for dissemination to new hosts. However, there are key gaps in knowledge regarding the mechanisms that regulate viral RNA packaging efficiency in bunyaviruses. Our studies investigating the mechanism of RNA packaging in RVFV revealed the presence of a direct interaction between the viral envelope glycoprotein, Gn, and the viral RNA segments in infected cells, for the first time in bunyaviruses. Furthermore, our data strongly indicate a critical role for the direct interaction between Gn and viral RNAs in determining the efficiency of incorporation of viral RNA segments into RVFV particles. Clarifying the fundamental mechanisms of RNA packaging in RVFV would be valuable for the development of antivirals and live-attenuated vaccines.


Asunto(s)
ARN Viral , Virus de la Fiebre del Valle del Rift/genética , Empaquetamiento del Genoma Viral , Secuencia de Empaquetamiento Viral , Virión/genética , Animales , Línea Celular , Chlorocebus aethiops , Ribonucleoproteínas/metabolismo , Células Vero , Proteínas del Envoltorio Viral/genética
3.
J Virol ; 89(21): 10970-81, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26311885

RESUMEN

UNLABELLED: The newly emerged Middle East respiratory syndrome coronavirus (MERS-CoV) and severe acute respiratory syndrome CoV (SARS-CoV) represent highly pathogenic human CoVs that share a property to inhibit host gene expression at the posttranscriptional level. Similar to the nonstructural protein 1 (nsp1) of SARS-CoV that inhibits host gene expression at the translational level, we report that MERS-CoV nsp1 also exhibits a conserved function to negatively regulate host gene expression by inhibiting host mRNA translation and inducing the degradation of host mRNAs. Furthermore, like SARS-CoV nsp1, the mRNA degradation activity of MERS-CoV nsp1, most probably triggered by its ability to induce an endonucleolytic RNA cleavage, was separable from its translation inhibitory function. Despite these functional similarities, MERS-CoV nsp1 used a strikingly different strategy that selectively targeted translationally competent host mRNAs for inhibition. While SARS-CoV nsp1 is localized exclusively in the cytoplasm and binds to the 40S ribosomal subunit to gain access to translating mRNAs, MERS-CoV nsp1 was distributed in both the nucleus and the cytoplasm and did not bind stably to the 40S subunit, suggesting a distinctly different mode of targeting translating mRNAs. Interestingly, consistent with this notion, MERS-CoV nsp1 selectively targeted mRNAs, which are transcribed in the nucleus and transported to the cytoplasm, for translation inhibition and mRNA degradation but spared exogenous mRNAs introduced directly into the cytoplasm or virus-like mRNAs that originate in the cytoplasm. Collectively, these data point toward a novel viral strategy wherein the cytoplasmic origin of MERS-CoV mRNAs facilitates their escape from the inhibitory effects of MERS-CoV nsp1. IMPORTANCE: Middle East respiratory syndrome coronavirus (MERS-CoV) is a highly pathogenic human CoV that emerged in Saudi Arabia in 2012. MERS-CoV has a zoonotic origin and poses a major threat to public health. However, little is known about the viral factors contributing to the high virulence of MERS-CoV. Many animal viruses, including CoVs, encode proteins that interfere with host gene expression, including those involved in antiviral immune responses, and these viral proteins are often major virulence factors. The nonstructural protein 1 (nsp1) of CoVs is one such protein that inhibits host gene expression and is a major virulence factor. This study presents evidence for a strategy used by MERS-CoV nsp1 to inhibit host gene expression that has not been described previously for any viral protein. The present study represents a meaningful step toward a better understanding of the factors and molecular mechanisms governing the virulence and pathogenesis of MERS-CoV.


Asunto(s)
Núcleo Celular/metabolismo , Regulación de la Expresión Génica/genética , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , ARN Mensajero/metabolismo , Proteínas no Estructurales Virales/metabolismo , Northern Blotting , Western Blotting , Citoplasma/metabolismo , Cartilla de ADN , Dipeptidil Peptidasa 4/metabolismo , Electroporación , Células HEK293 , Humanos , Microscopía Confocal , Coronavirus del Síndrome Respiratorio de Oriente Medio/metabolismo , Plásmidos/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
J Virol ; 87(1): 676-82, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23097454

RESUMEN

The NSm nonstructural protein of Rift Valley fever virus (family Bunyaviridae, genus Phlebovirus) has an antiapoptotic function and affects viral pathogenesis. We found that NSm integrates into the mitochondrial outer membrane and that the protein's N terminus is exposed to the cytoplasm. The C-terminal region of NSm, which contains a basic amino acid cluster and a putative transmembrane domain, targeted the protein to the mitochondrial outer membrane and exerted antiapoptotic function.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Membranas Mitocondriales/metabolismo , Transporte de Proteínas , Virus de la Fiebre del Valle del Rift/patogenicidad , Proteínas no Estructurales Virales/metabolismo , Animales , Línea Celular , Interacciones Huésped-Patógeno , Humanos
5.
Proc Natl Acad Sci U S A ; 108(2): 804-9, 2011 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-21187405

RESUMEN

The Bunyaviridae family includes pathogens of medical and veterinary importance. Rift Valley fever virus (RVFV), a member in the Phlebovirus genus of the family Bunyaviridae, is endemic to sub-Saharan Africa and causes a mosquito-borne disease in ruminants and humans. Viruses in the family Bunyaviridae carry a tripartite, single-stranded, negative-sense RNA genome composed of L, M, and S RNAs. Little is known about how the three genomic RNA segments are copackaged to generate infectious bunyaviruses. We explored the mechanism that governs the copackaging of the three genomic RNAs into RVFV particles. The expression of viral structural proteins along with replicating S and M RNAs resulted in the copackaging of both RNAs into RVFV-like particles, while replacing M RNA with M1 RNA, lacking a part of the M RNA 5' UTR, abrogated the RNA copackaging. L RNA was efficiently packaged into virus particles released from cells supporting the replication of L, M, and S RNAs, and replacing M RNA with M1 RNA abolished the packaging of L RNA. Detailed analyses using various combinations of replicating viral RNAs suggest that M RNA alone or a coordinated function of M and S RNAs exerted efficient L RNA packaging either directly or indirectly. Collectively, these data are consistent with the possibility that specific intermolecular interactions among the three viral RNAs drive the copackaging of these RNAs to produce infectious RVFV.


Asunto(s)
Genoma Viral , Fiebres Hemorrágicas Virales/virología , ARN Viral/genética , ARN/genética , Fiebre del Valle del Rift/genética , Virus de la Fiebre del Valle del Rift/genética , Regiones no Traducidas 5' , Animales , Northern Blotting , Western Blotting , Chlorocebus aethiops , Eliminación de Gen , Modelos Genéticos , Mutación , Células Vero
6.
J Virol ; 86(7): 4034-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22278239

RESUMEN

We characterized the RNA elements involved in the packaging of Rift Valley fever virus RNA genome segments, L, M, and S. The 5'-terminal 25 nucleotides of each RNA segment were equally competent for RNA packaging and carried an RNA packaging signal, which overlapped with the RNA replication signal. Only the deletion mutants of L RNA, but not full-length L RNA, were efficiently packaged, implying the possible requirement of RNA compaction for L RNA packaging.


Asunto(s)
Genoma Viral , ARN Viral/genética , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/genética , Ensamble de Virus , Secuencia de Bases , Línea Celular , Humanos , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Virus de la Fiebre del Valle del Rift/fisiología , Replicación Viral
7.
Front Cell Infect Microbiol ; 13: 1132757, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36875526

RESUMEN

Rift Valley fever virus (RVFV), a bunyavirus, has a single-stranded, negative-sense tri-segmented RNA genome, consisting of L, M and S RNAs. An infectious virion carries two envelope glycoproteins, Gn and Gc, along with ribonucleoprotein complexes composed of encapsidated viral RNA segments. The antigenomic S RNA, which serves as the template of the mRNA encoding a nonstructural protein, NSs, an interferon antagonist, is also efficiently packaged into RVFV particles. An interaction between Gn and viral ribonucleoprotein complexes, including the direct binding of Gn to viral RNAs, drives viral RNA packaging into RVFV particles. To understand the mechanism of efficient antigenomic S RNA packaging in RVFV, we identified the regions in viral RNAs that directly interact with Gn by performing UV-crosslinking and immunoprecipitation of RVFV-infected cell lysates with anti-Gn antibody followed by high-throughput sequencing analysis (CLIP-seq analysis). Our data suggested the presence of multiple Gn-binding sites in RVFV RNAs, including a prominent Gn-binding site within the 3' noncoding region of the antigenomic S RNA. We found that the efficient packaging of antigenomic S RNA was abrogated in a RVFV mutant lacking a part of this prominent Gn-binding site within the 3' noncoding region. Also, the mutant RVFV, but not the parental RVFV, triggered the early induction of interferon-ß mRNA expression after infection. These data suggest that the direct binding of Gn to the RNA element within the 3' noncoding region of the antigenomic S RNA promoted the efficient packaging of antigenomic S RNA into virions. Furthermore, the efficient packaging of antigenomic S RNA into RVFV particles, driven by the RNA element, facilitated the synthesis of viral mRNA encoding NSs immediately after infection, resulting in the suppression of interferon-ß mRNA expression.


Asunto(s)
Virus de la Fiebre del Valle del Rift , Animales , ARN Viral , ARN Mensajero , Interferón beta , Ribonucleoproteínas
8.
PLoS Negl Trop Dis ; 15(9): e0009785, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34516560

RESUMEN

Rift Valley fever virus (RVFV) is a mosquito-borne bunyavirus with a wide host range including ruminants and humans. RVFV outbreaks have had devastating effects on public health and the livestock industry in African countries. However, there is no approved RVFV vaccine for human use in non-endemic countries and no FDA-approved antiviral drug for RVFV treatment. The RVFV 78kDa protein (P78), which is a membrane glycoprotein, plays a role in virus dissemination in the mosquito host, but its biological role in mammalian hosts remains unknown. We generated an attenuated RVFV MP-12 strain-derived P78-High virus and a virulent ZH501 strain-derived ZH501-P78-High virus, both of which expressed a higher level of P78 and carried higher levels of P78 in the virion compared to their parental viruses. We also generated another MP-12-derived mutant virus (P78-KO virus) that does not express P78. MP-12 and P78-KO virus replicated to similar levels in fibroblast cell lines and Huh7 cells, while P78-High virus replicated better than MP-12 in Vero E6 cells, fibroblast cell lines, and Huh7 cells. Notably, P78-High virus and P78-KO virus replicated less efficiently and more efficiently, respectively, than MP-12 in macrophage cell lines. ZH501-P78-High virus also replicated poorly in macrophage cell lines. Our data further suggest that inefficient binding of P78-High virus to the cells led to inefficient virus internalization, low virus infectivity and reduced virus replication in a macrophage cell line. P78-High virus and P78-KO virus showed lower and higher virulence than MP-12, respectively, in young mice. ZH501-P78-High virus also exhibited lower virulence than ZH501 in mice. These data suggest that high levels of P78 expression attenuate RVFV virulence by preventing efficient virus replication in macrophages. Genetic alteration leading to increased P78 expression may serve as a novel strategy for the attenuation of RVFV virulence and generation of safe RVFV vaccines.


Asunto(s)
Macrófagos/virología , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/fisiología , Proteínas del Envoltorio Viral/metabolismo , Replicación Viral/fisiología , Animales , Ratones , Virus de la Fiebre del Valle del Rift/patogenicidad , Proteínas del Envoltorio Viral/genética , Virulencia
9.
J Virol ; 83(24): 12779-89, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19812169

RESUMEN

Rift Valley fever virus (RVFV) (genus Phlebovirus, family Bunyaviridae) causes mosquito-borne epidemic diseases in humans and livestock. The virus carries three RNA segments, L, M, and S, of negative or ambisense polarity. L protein, an RNA-dependent RNA polymerase, encoded in the L segment, and N protein, encoded in the S segment, exert viral RNA replication and transcription. Coexpression of N, hemagglutinin (HA)-tagged L, and viral minigenome resulted in minigenome replication and transcription, a finding that demonstrated HA-tagged L was biologically active. Likewise L tagged with green fluorescent protein (GFP) was biologically competent. Coimmunoprecipitation analysis using extracts from cells coexpressing HA-tagged L and GFP-tagged L showed the formation of an L oligomer. Bimolecular fluorescence complementation analysis and coimmunoprecipitation studies demonstrated the formation of an intermolecular L-L interaction through its N-terminal and C-terminal regions and also suggested an intramolecular association between the N-terminal and C-terminal regions of L protein. A biologically inactive L mutant, in which the conserved signature SDD motif was replaced by the amino acid residues GNN, exhibited a dominant negative phenotype when coexpressed with wild-type L in the minigenome assay system. Expression of this mutant L also inhibited viral gene expression in virus-infected cells. These data provided compelling evidence for the importance of oligomerization of RVFV L protein for its polymerase activity.


Asunto(s)
ARN Polimerasa Dependiente del ARN/química , Virus de la Fiebre del Valle del Rift/química , Proteínas Virales/química , Células Cultivadas , Humanos , Mutación , ARN Polimerasa Dependiente del ARN/fisiología
10.
J Virol Methods ; 272: 113701, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31315022

RESUMEN

Rift Valley Fever phlebovirus (RVFV), genus Phlebovirus, family Phenuiviridae, order Bunyavirales, has a single-stranded, negative-sense RNA genome, consisting of L, M and S segments. Here, we report the establishment of a strand-specific, quantitative reverse transcription (RT)-PCR assay system that can selectively distinguish between the genomic and antigenomic RNAs of each of the three viral RNA segments produced in RVFV-infected cells. To circumvent the obstacle of primer-independent cDNA synthesis during RT, we used a tagged, strand-specific RT primer, carrying a non-viral 'tag' sequence at the 5' end, which ensured the strand-specificity through the selective amplification of only the tagged cDNA in the real-time PCR assay. We used this assay system to examine the kinetics of intracellular accumulation of genomic and antigenomic viral RNAs in mammalian cells infected with the MP-12 strain of RVFV. The genomic RNA copy numbers, for all three viral RNA segments, were higher than that of their corresponding antigenomic RNAs throughout the time-course of infection, with a notable exception, wherein the M segment genomic and antigenomic RNAs exhibited similar copy numbers at specific times post-infection. Overall, this assay system could be a useful tool to gain an insight into the mechanisms of RNA replication and packaging in RVFV.


Asunto(s)
Genómica/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Fiebre del Valle del Rift/diagnóstico , Virus de la Fiebre del Valle del Rift/genética , Virus de la Fiebre del Valle del Rift/aislamiento & purificación , Animales , Línea Celular , Chlorocebus aethiops , ADN Complementario , Humanos , Cinética , Técnicas de Diagnóstico Molecular , ARN Viral/genética , ARN Viral/aislamiento & purificación , Fiebre del Valle del Rift/virología , Sensibilidad y Especificidad , Células Vero , Replicación Viral
11.
J Vet Med Sci ; 70(4): 407-10, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18460838

RESUMEN

Susceptibility of DT40 cells to pathogenic field strains of infectious bursal disease virus (IBDV) including very virulent and classical virulent strains were studied. After the first and second passage of the virus in DT40 cells, IBDV-specific antigen was readily detected in DT40 cells inoculated with the pathogenic field strain infected bursal homogenates. Nucleotide sequence analysis in the VP2 hypervariable domain, which is critical for the virulence of IBDV, revealed no common amino acid substitutions among the pathogenic IBDVs in accordance with the propagation in DT40 cells. These results indicate that DT40 cells are a useful tool for rapid isolation of pathogenic field strains and successive in vitro analysis of IBDV.


Asunto(s)
Pollos , Virus de la Enfermedad Infecciosa de la Bolsa/fisiología , Linfoma , Cultivo de Virus/veterinaria , Animales , Línea Celular Tumoral , Factores de Tiempo , Replicación Viral/fisiología
12.
Sci Rep ; 8(1): 17097, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30459418

RESUMEN

Rift Valley fever phlebovirus (RVFV) is a pathogen of Rift Valley fever, which is a mosquito-borne zoonotic disease for domestic livestock and humans in African countries. Currently, no approved vaccine is available for use in non-endemic areas. The MP-12 strain is so far the best live attenuated RVFV vaccine candidate because of its good protective efficacy in animal models. However, there are safety concerns for use of MP-12 in humans. We previously developed a single-cycle replicable MP-12 (scMP-12) which lacks NSs gene and undergoes only a single round of viral replication because of its impaired ability to induce membrane-membrane fusion. In the present study, we generated an scMP-12 mutant (scMP-12-mutNSs) carrying a mutant NSs, which degrades double-stranded RNA-dependent protein kinase R but does not inhibit host transcription. Immunization of mice with a single dose (105 PFU) of scMP-12-mutNSs elicited RVFV neutralizing antibodies and high titers of anti-N IgG production and fully protected the mice from lethal wild-type RVFV challenge. Immunogenicity and protective efficacy of scMP-12-mutNSs were better than scMP-12, demonstrating that scMP-12-mutNSs is a more efficacious vaccine candidate than scMP-12. Furthermore, our data suggested that RVFV vaccine efficacy can be improved by using this specific NSs mutant.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Mutación , Fiebre del Valle del Rift/prevención & control , Virus de la Fiebre del Valle del Rift/patogenicidad , Vacunas Atenuadas/administración & dosificación , Proteínas no Estructurales Virales/genética , Vacunas Virales/administración & dosificación , África , Animales , Femenino , Ratones , Fiebre del Valle del Rift/inmunología , Fiebre del Valle del Rift/virología , Vacunación , Replicación Viral
13.
J Vet Med Sci ; 69(7): 733-8, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17675805

RESUMEN

Bursae of Fabricius were collected from 20 chickens diagnosed with infectious bursal disease virus (IBDV) infection from 15 prefectures in 1993 to 2004. Here we report the nucleotide sequence analysis of VP2 hypervariable domain of IBDV genome detected by reverse transcription-polymerase chain reaction from these samples. Ten sequences derived from 10 prefectures in 1996 to 2003 were of the classical type and other 10 sequences derived from 6 prefectures in 1993 to 2004 were of the highly virulent type. Of the classical type sequences, 9 sequences were closely related to the sequence of classical attenuated vaccines used in Japan. Furthermore, two were identical to the sequence of B-Chi5 which represents Vaccine B passaged 5 times in chickens and was reported to be reverted the virulence during the passages. The 10 highly virulent type sequences were classified into four sequences, none of which had been previously detected in Japan. However, the deduced amino acid sequences were identical to each other and to the sequences of highly virulent IBDVs previously detected in Japan. The most common nucleotide sequences, which accounted for 6 of the sequences, were identical to 34 highly virulent type sequences detected in various countries in BLAST search. This is the first report of detection of the sequence in Japan which is identical to highly virulent strains detected in other countries. These findings show the prevalence of classical IBDVs closely related to the attenuated vaccines and highly virulent IBDVs derived from other countries throughout Japan since 1993.


Asunto(s)
Infecciones por Birnaviridae/veterinaria , Pollos , Brotes de Enfermedades/veterinaria , Virus de la Enfermedad Infecciosa de la Bolsa/genética , Enfermedades de las Aves de Corral/virología , Proteínas Estructurales Virales/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Infecciones por Birnaviridae/epidemiología , Infecciones por Birnaviridae/virología , Virus de la Enfermedad Infecciosa de la Bolsa/patogenicidad , Japón/epidemiología , Datos de Secuencia Molecular , Filogenia , Enfermedades de las Aves de Corral/epidemiología , Estructura Terciaria de Proteína , ARN Viral/química , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Alineación de Secuencia , Proteínas Estructurales Virales/química , Virulencia
14.
Avian Dis ; 50(4): 556-60, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17274294

RESUMEN

A practical sampling method for bursal tissue using ordinary paper for molecular diagnosis of infectious bursal disease (IBD) was established. IBD virus-infected bursa was directly smeared on chromatography paper, filter paper, or stationery copy paper and was then fixed with absolute ethanol, Tris-HCl-saturated phenol, or phenol:chloroform:isoamyl alcohol (25:24:1). Flinders Technology Associates (FTA) card, which is designed for the collection of biological samples for molecular detection, was also used. After storage at 37 C for up to 30 days, total RNA directly extracted from the tissue fixed on the papers and FTA card were subjected to reverse transcriptase-polymerase chain reaction (RT-PCR) for detection of IBD virus (IBDV) RNA. In addition, the ability of each chemical used in the fixation and the FTA card to inactivate IBDV was evaluated. Regardless of the paper quality, storage period, and fixation method, IBDV RNA was consistently detected in all of the samples. IBDV in the bursal tissue was inactivated with phenol but not with ethanol or the unknown chemicals in FTA card. These results show that ordinary papers sustain the viral RNA, as does FTA card, but phenol fixation is superior to FTA card in inactivating IBDV. The new sampling method using ordinary paper with phenol fixation is safe, inexpensive, simple, and easy, and is thus suitable for conducting a global survey of IBD even where laboratory resources are limited. This practical method should contribute to the control of IBD worldwide.


Asunto(s)
Infecciones por Birnaviridae/veterinaria , Pollos/virología , Virus de la Enfermedad Infecciosa de la Bolsa/aislamiento & purificación , ARN Viral/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Animales , Infecciones por Birnaviridae/diagnóstico , Infecciones por Birnaviridae/virología , Bolsa de Fabricio/virología , Papel , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Organismos Libres de Patógenos Específicos
15.
Methods Mol Biol ; 1403: 187-206, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27076131

RESUMEN

Rift Valley fever virus (RVFV) (genus Phlebovirus, family Bunyaviridae) is an arbovirus that causes severe disease in humans and livestock in sub-Saharan African countries. The virus carries a tripartite, single-stranded, and negative-sense RNA genome, designated as L, M, and S RNAs. RVFV spread can be prevented by the effective vaccination of animals and humans. Although the MP-12 strain of RVFV is a live attenuated vaccine candidate, MP-12 showed neuroinvasiveness and neurovirulence in young mice and immunodeficiency mice. Hence, there is a concern for the use of MP-12 to certain individuals, especially those that are immunocompromised. To improve MP-12 safety, we have generated a single-cycle, replicable MP-12 (scMP-12), which carries L RNA, S RNA encoding green fluorescent protein in place of a viral nonstructural protein NSs, and an M RNA encoding a mutant envelope protein lacking an endoplasmic reticulum retrieval signal and defective for membrane fusion function. The scMP-12 undergoes efficient amplification in the Vero-G cell line, which is a Vero cell line stably expressing viral envelope proteins, while it undergoes single-cycle replication in naïve cells and completely lacks neurovirulence in suckling mice after intracranial inoculation. A single-dose vaccination of mice with scMP-12 confers protective immunity. Thus, scMP-12 represents a new, promising RVF vaccine candidate. Here we describe protocols for scMP-12 generation by using a reverse genetics system, establishment of Vero-G cells, and titration of scMP-12 in Vero-G cells.


Asunto(s)
Genética Inversa/métodos , Vacunas Virales/genética , Animales , Chlorocebus aethiops , Humanos , Virus de la Fiebre del Valle del Rift/inmunología , Células Vero , Vacunas Virales/inmunología
16.
Virus Res ; 216: 55-65, 2016 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-26022573

RESUMEN

Rift Valley fever virus (RVFV) is an arbovirus circulating between ruminants and mosquitoes to maintain its enzootic cycle. Humans are infected with RVFV through mosquito bites or direct contact with materials of infected animals. The virus causes Rift Valley fever (RVF), which was first recognized in the Great Rift Valley of Kenya in 1931. RVF is characterized by a febrile illness resulting in a high rate of abortions in ruminants and an acute febrile illness, followed by fatal hemorrhagic fever and encephalitis in humans. Initially, the virus was restricted to the eastern region of Africa, but the disease has now spread to southern and western Africa, as well as outside of the African continent, e.g., Madagascar, Saudi Arabia and Yemen. There is a serious concern that the virus may spread to other areas, such as North America and Europe. As vaccination is an effective tool to control RVFV epidemics, formalin-inactivated vaccines and live-attenuated RVFV vaccines have been used in endemic areas. The formalin-inactivated vaccines require boosters for effective protection, whereas the live-attenuated vaccines enable the induction of protective immunity by a single vaccination. However, the use of live-attenuated RVFV vaccines for large human populations having a varied health status is of concern, because of these vaccines' residual neuro-invasiveness and neurovirulence. Recently, novel vaccine candidates have been developed using replication-defective RVFV that can undergo only a single round of replication in infected cells. The single-cycle replicable RVFV does not cause systemic infection in immunized hosts, but enables the conferring of protective immunity. This review summarizes the properties of various RVFV vaccines and recent progress on the development of the single-cycle replicable RVFV vaccines.


Asunto(s)
Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/inmunología , Vacunas Virales/inmunología , Replicación Viral , Animales , Humanos , Fiebre del Valle del Rift/inmunología , Fiebre del Valle del Rift/prevención & control , Virus de la Fiebre del Valle del Rift/genética , Virus de la Fiebre del Valle del Rift/fisiología , Vacunas Virales/genética
17.
PLoS Negl Trop Dis ; 10(10): e0005047, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27711108

RESUMEN

Rift Valley fever virus (RVFV), a member of the genus Phlebovirus within the family Bunyaviridae, causes periodic outbreaks in livestocks and humans in countries of the African continent and Middle East. RVFV NSs protein, a nonstructural protein, is a major virulence factor that exhibits several important biological properties. These include suppression of general transcription, inhibition of IFN-ß promoter induction and degradation of double-stranded RNA-dependent protein kinase R. Although each of these biological functions of NSs are considered important for countering the antiviral response in the host, the individual contributions of these functions towards RVFV virulence remains unclear. To examine this, we generated two RVFV MP-12 strain-derived mutant viruses. Each carried mutations in NSs that specifically targeted its general transcription inhibition function without affecting its ability to degrade PKR and inhibit IFN-ß promoter induction, through its interaction with Sin3-associated protein 30, a part of the repressor complex at the IFN-ß promoter. Using these mutant viruses, we have dissected the transcription inhibition function of NSs and examined its importance in RVFV virulence. Both NSs mutant viruses exhibited a differentially impaired ability to inhibit host transcription when compared with MP-12. It has been reported that NSs suppresses general transcription by interfering with the formation of the transcription factor IIH complex, through the degradation of the p62 subunit and sequestration of the p44 subunit. Our study results lead us to suggest that the ability of NSs to induce p62 degradation is the major contributor to its general transcription inhibition property, whereas its interaction with p44 may not play a significant role in this function. Importantly, RVFV MP-12-NSs mutant viruses with an impaired general transcription inhibition function showed a reduced cytotoxicity in cell culture and attenuated virulence in young mice, compared with its parental virus MP-12, highlighting the contribution of NSs-mediated general transcription inhibition towards RVFV virulence.


Asunto(s)
Virus de la Fiebre del Valle del Rift/genética , Virus de la Fiebre del Valle del Rift/patogenicidad , Factores de Transcripción/metabolismo , Transcripción Genética , Proteínas no Estructurales Virales/fisiología , Factores de Virulencia/fisiología , Animales , Chlorocebus aethiops , Interacciones Huésped-Patógeno , Humanos , Interferón beta/genética , Ratones , Mutación , Regiones Promotoras Genéticas , Fiebre del Valle del Rift/virología , Factores de Transcripción/genética , Células Vero , Proteínas no Estructurales Virales/genética , Factores de Virulencia/genética
18.
Antiviral Res ; 127: 79-89, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26801627

RESUMEN

Rift Valley fever virus (RVFV), genus Phlebovirus family Bunyaviridae, is an arthropod-borne virus endemic throughout sub-Saharan Africa. Recent outbreaks have resulted in cyclic epidemics with an increasing geographic footprint, devastating both livestock and human populations. Despite being recognized as an emerging threat, relatively little is known about the virulence mechanisms and host interactions of RVFV. To date there are no FDA approved therapeutics or vaccines for RVF and there is an urgent need for their development. The Ser/Thr protein phosphatase 1 (PP1) has previously been shown to play a significant role in the replication of several viruses. Here we demonstrate for the first time that PP1 plays a prominent role in RVFV replication early on during the viral life cycle. Both siRNA knockdown of PP1α and a novel PP1-targeting small molecule compound 1E7-03, resulted in decreased viral titers across several cell lines. Deregulation of PP1 was found to inhibit viral RNA production, potentially through the disruption of viral RNA transcript/protein interactions, and indicates a potential link between PP1α and the viral L polymerase and nucleoprotein. These results indicate that PP1 activity is important for RVFV replication early on during the viral life cycle and may prove an attractive therapeutic target.


Asunto(s)
Antivirales/farmacología , Replicación del ADN/fisiología , Indoles/farmacología , Proteína Fosfatasa 1/metabolismo , Virus de la Fiebre del Valle del Rift/enzimología , Virus de la Fiebre del Valle del Rift/fisiología , Urea/análogos & derivados , Replicación Viral/fisiología , Animales , Antivirales/química , Antivirales/uso terapéutico , Línea Celular , Replicación del ADN/efectos de los fármacos , Genoma Viral/efectos de los fármacos , Interacciones Huésped-Patógeno , Humanos , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteína Fosfatasa 1/antagonistas & inhibidores , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , ARN Viral/biosíntesis , ARN Viral/efectos de los fármacos , Fiebre del Valle del Rift/tratamiento farmacológico , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Virus de la Fiebre del Valle del Rift/genética , Urea/farmacología , Células Vero , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/metabolismo , Virulencia , Replicación Viral/efectos de los fármacos
19.
J Innate Immun ; 7(5): 450-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25766761

RESUMEN

Rift Valley fever virus (RVFV) belongs to the genus Phlebovirus, family Bunyaviridae, and carries single-stranded tripartite RNA segments. The virus is transmitted by mosquitoes and has caused large outbreaks among ruminants and humans in sub-Saharan African and Middle East countries. The disease is characterized by a sudden onset of fever, headache, muscle pain, joint pain, photophobia, and weakness. In most cases, patients recover from the disease after a period of weeks, but some also develop retinal or macular changes, which result in vision impairment that lasts for an undefined period of time, and severe disease, characterized by hemorrhagic fever or encephalitis. The virus also causes febrile illness resulting in a high rate of spontaneous abortions in ruminants. The handling of wild-type RVFV requires high-containment facilities, including biosafety level 4 or enhanced biosafety level 3 laboratories. Nonetheless, studies clarifying the mechanisms of the RVFV-induced diseases and preventing them are areas of active research throughout the world. By primarily referring to recent studies using several animal model systems, protein expression systems, and specific mutant viruses, this review describes the current knowledge about the mechanisms of pathogenesis of RVF and biological functions of various viral proteins that affect RVFV pathogenicity.


Asunto(s)
Fiebre del Valle del Rift/inmunología , Virus de la Fiebre del Valle del Rift/inmunología , Proteínas Virales/metabolismo , Animales , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Humanos
20.
PLoS Negl Trop Dis ; 8(3): e2746, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24651859

RESUMEN

Rift Valley fever virus (RVFV) (genus Phlebovirus, family Bunyaviridae) is an arbovirus that causes severe disease in humans and livestock in sub-Saharan African countries. Although the MP-12 strain of RVFV is a live attenuated vaccine candidate, neuroinvasiveness and neurovirulence of MP-12 in mice may be a concern when vaccinating certain individuals, especially those that are immunocompromised. We have developed a novel, single-cycle replicable MP-12 (scMP-12), which carries an L RNA, M RNA mutant encoding a mutant envelope protein lacking an endoplasmic reticulum retrieval signal and defective for membrane fusion function, and S RNA encoding N protein and green fluorescent protein. The scMP-12 underwent efficient amplification, then formed plaques and retained the introduced mutation after serial passages in a cell line stably expressing viral envelope proteins. However, inoculation of the scMP-12 into naïve cells resulted in a single round of viral replication, and production of low levels of noninfectious virus-like particles. Intracranial inoculation of scMP-12 into suckling mice did not cause clinical signs or death, a finding which demonstrated that the scMP-12 lacked neurovirulence. Mice immunized with a single dose of scMP-12 produced neutralizing antibodies, whose titers were higher than in mice immunized with replicon particles carrying L RNA and S RNA encoding N protein and green fluorescent protein. Moreover, 90% of the scMP-12-immunized mice were protected from wild-type RVFV challenge by efficiently suppressing viremia and replication of the challenge virus in the liver and the spleen. These data demonstrated that scMP-12 is a safe and immunogenic RVFV vaccine candidate.


Asunto(s)
Fiebre del Valle del Rift/prevención & control , Virus de la Fiebre del Valle del Rift/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Modelos Animales de Enfermedad , Femenino , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Virus de la Fiebre del Valle del Rift/genética , Virus de la Fiebre del Valle del Rift/fisiología , Análisis de Supervivencia , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/efectos adversos , Vacunas Virales/genética , Internalización del Virus , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA