Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Org Lett ; 23(24): 9649-9653, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34878280

RESUMO

Miyaura borylation, that is, palladium-catalyzed cross-coupling between bromoarenes and diboron, offers a versatile method for preparing arylboronates; however, a costly and inaccessible catalyst has been required for synthesizing highly congested arylboronates with the method. Here the Pd(OAc)2-tri(4-methoxyphenyl)phosphine catalyst was found to work as an efficient catalyst for the sterically demanding borylation. A broad range of o,o-disubstituted bromoarenes were converted into the corresponding arylboronates in high yields by using the palladium catalyst with Cs2CO3 in EtOAc at 80 °C.

2.
Proc Natl Acad Sci U S A ; 111(20): 7367-72, 2014 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-24799716

RESUMO

Sterile alpha motif domain containing protein 4 (Samd4) is an RNA binding protein that mediates translational repression. We identified a Samd4 missense mutation, designated supermodel, that caused leanness and kyphosis associated with myopathy and adipocyte defects in C57BL/6J mice. The supermodel mutation protected homozygous mice from high fat diet-induced obesity, likely by promoting enhanced energy expenditure through uncoupled mitochondrial respiration. Glucose tolerance was impaired due to diminished insulin release in homozygous mutant mice. The defects of metabolism in supermodel mice may be explained by dysregulated mechanistic target of rapamycin complex 1 (mTORC1) signaling, evidenced by hypophosphorylation of 4E-BP1 and S6 in muscle and adipose tissues of homozygous mice. Samd4 may interface with mTORC1 signaling through an interaction with 14-3-3 proteins and with Akt, which phosphorylates Samd4 in vitro.


Assuntos
Complexos Multiproteicos/metabolismo , Mutação , Proteínas Repressoras/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Proteínas 14-3-3/metabolismo , Adipócitos/citologia , Motivos de Aminoácidos , Animais , Composição Corporal , Feminino , Glucose/metabolismo , Teste de Tolerância a Glucose , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese , Fenótipo , Fosforilação , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/fisiologia
3.
J Virol ; 87(1): 676-82, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23097454

RESUMO

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.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Membranas Mitocondriais/metabolismo , Transporte Proteico , Vírus da Febre do Vale do Rift/patogenicidade , Proteínas não Estruturais Virais/metabolismo , Animais , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos
4.
BMC Res Notes ; 5: 577, 2012 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-23095377

RESUMO

BACKGROUND: We present a compendium of N-ethyl-N-nitrosourea (ENU)-induced mouse mutations, identified in our laboratory over a period of 10 years either on the basis of phenotype or whole genome and/or whole exome sequencing, and archived in the Mutagenetix database. Our purpose is threefold: 1) to formally describe many point mutations, including those that were not previously disclosed in peer-reviewed publications; 2) to assess the characteristics of these mutations; and 3) to estimate the likelihood that a missense mutation induced by ENU will create a detectable phenotype. FINDINGS: In the context of an ENU mutagenesis program for C57BL/6J mice, a total of 185 phenotypes were tracked to mutations in 129 genes. In addition, 402 incidental mutations were identified and predicted to affect 390 genes. As previously reported, ENU shows strand asymmetry in its induction of mutations, particularly favoring T to A rather than A to T in the sense strand of coding regions and splice junctions. Some amino acid substitutions are far more likely to be damaging than others, and some are far more likely to be observed. Indeed, from among a total of 494 non-synonymous coding mutations, ENU was observed to create only 114 of the 182 possible amino acid substitutions that single base changes can achieve. Based on differences in overt null allele frequencies observed in phenotypic vs. non-phenotypic mutation sets, we infer that ENU-induced missense mutations create detectable phenotype only about 1 in 4.7 times. While the remaining mutations may not be functionally neutral, they are, on average, beneath the limits of detection of the phenotypic assays we applied. CONCLUSIONS: Collectively, these mutations add to our understanding of the chemical specificity of ENU, the types of amino acid substitutions it creates, and its efficiency in causing phenovariance. Our data support the validity of computational algorithms for the prediction of damage caused by amino acid substitutions, and may lead to refined predictions as to whether specific amino acid changes are responsible for observed phenotypes. These data form the basis for closer in silico estimations of the number of genes mutated to a state of phenovariance by ENU within a population of G3 mice.


Assuntos
Etilnitrosoureia/toxicidade , Mutagênicos/toxicidade , Mutação , Alelos , Animais , Bases de Dados Genéticas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
J Virol ; 86(3): 1802-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22114338

RESUMO

The downregulation of translation through eIF2α phosphorylation is a cellular response to diverse stresses, including viral infection, and is mediated by the GCN2 kinase, protein kinase R (PKR), protein kinase-like endoplasmic reticulum kinase (PERK), and heme-regulated inhibitor kinase (HRI). Although PKR plays a major role in defense against viruses, other eIF2α kinases also may respond to viral infection and contribute to the shutdown of protein synthesis. Here we describe the recessive, loss-of-function mutation atchoum (atc) in Eif2ak4, encoding GCN2, which increased susceptibility to infection by the double-stranded DNA viruses mouse cytomegalovirus (MCMV) and human adenovirus. This mutation was identified by screening macrophages isolated from mice carrying N-ethyl-N-nitrosourea (ENU)-induced mutations. Cells from Eif2ak4(atc/atc) mice failed to phosphorylate eIF2α in response to MCMV. Importantly, homozygous Eif2ak4(atc) mice showed a modest increase in susceptibility to MCMV infection, demonstrating that translational arrest dependent on GCN2 contributes to the antiviral response in vivo.


Assuntos
Infecções por Vírus de DNA/genética , Predisposição Genética para Doença , Mutação , Proteínas Serina-Treonina Quinases/genética , Animais , Sequência de Bases , Primers do DNA , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Proc Natl Acad Sci U S A ; 108(29): 12024-9, 2011 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-21719711

RESUMO

The effects of the cellular environment on innate immunity remain poorly characterized. Here, we show that in Drosophila ATP-sensitive potassium channels (K(ATP)) mediate resistance to a cardiotropic RNA virus, Flock House virus (FHV). FHV viral load in the heart rapidly increases in K(ATP) mutant flies, leading to increased viremia and accelerated death. The effect of K(ATP) channels is dependent on the RNA interference genes Dcr-2, AGO2, and r2d2, indicating that an activity associated with this potassium channel participates in this antiviral pathway in Drosophila. Flies treated with the K(ATP) agonist drug pinacidil are protected against FHV infection, thus demonstrating the importance of this regulation of innate immunity by the cellular environment in the heart. In mice, the Coxsackievirus B3 replicates to higher titers in the hearts of mayday mutant animals, which are deficient in the Kir6.1 subunit of K(ATP) channels, than in controls. Together, our data suggest that K(ATP) channel deregulation can have a critical impact on innate antiviral immunity in the heart.


Assuntos
Drosophila/imunologia , Drosophila/virologia , Coração/virologia , Imunidade Inata/imunologia , Canais KATP/metabolismo , Nodaviridae/imunologia , Animais , Células HeLa , Humanos , Immunoblotting , Canais KATP/agonistas , Canais KATP/genética , Camundongos , Camundongos Endogâmicos C57BL , Nodaviridae/efeitos dos fármacos , Pinacidil/farmacologia , Interferência de RNA/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tolbutamida , Carga Viral/imunologia , Viremia
7.
Genetics ; 186(4): 1139-46, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20923982

RESUMO

Phenovariance may be obscured when genetic mapping is performed using highly divergent strains, and closely similar strains are preferred if adequate marker density can be established. We sequenced the C57BL/10J mouse genome using the Applied Biosystems SOLiD platform and here describe a genome-wide panel of informative markers that permits the mapping of mutations induced on the closely related C57BL/6J background by outcrossing to C57BL/10J, and backcrossing or intercrossing. The panel consists of 127 single nucleotide polymorphisms validated by capillary sequencing: 124 spaced at ∼20-Mb intervals across the 19 autosomes, and three markers on the X chromosome. We determined the genetic relationship between four C57BL-derived substrains and used the panel to map two N-ethyl-N-nitrosourea (ENU)-induced mutations responsible for visible phenotypes in C57BL/6J mice through bulk segregation analysis. Capillary sequencing, with computation of relative chromatogram peak heights, was used to determine the proportion of alleles from each strain at each marker.


Assuntos
Genoma/genética , Genômica/métodos , Mutação , Análise de Sequência de DNA/métodos , Alelos , Animais , Cromossomos , Marcadores Genéticos , Camundongos , Camundongos Endogâmicos , Mutagênese/efeitos dos fármacos , Mutação/efeitos dos fármacos , Mutação/genética , Fenótipo , Polimorfismo de Nucleotídeo Único , Especificidade da Espécie , Cromossomo X
8.
PLoS One ; 5(4): e9986, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20376320

RESUMO

BACKGROUND: Rift Valley fever virus (RVFV), a member of the genus Phlebovirus within the family Bunyaviridae, is a negative-stranded RNA virus with a tripartite genome. RVFV is transmitted by mosquitoes and causes fever and severe hemorrhagic illness among humans, while in livestock it causes fever and high abortion rates. METHODOLOGY/PRINCIPAL FINDINGS: Sequence analysis showed that a wild-type RVFV ZH501 preparation consisted of two major viral subpopulations, with a single nucleotide heterogeneity at nucleotide 847 of M segment (M847); one had a G residue at M847 encoding glycine in a major viral envelope Gn protein, while the other carried A residue encoding glutamic acid at the corresponding site. Two ZH501-derived viruses, rZH501-M847-G and rZH501-M847-A, carried identical genomic sequences, except that the former and the latter had G and A, respectively, at M847 were recovered by using a reverse genetics system. Intraperitoneal inoculation of rZH501-M847-A into mice caused a rapid and efficient viral accumulation in the sera, livers, spleens, kidneys and brains, and killed most of the mice within 8 days, whereas rZH501-M847-G caused low viremia titers, did not replicate as efficiently as did rZH501-M847-A in these organs, and had attenuated virulence to mice. Remarkably, as early as 2 days postinfection with rZH501-M847-G, the viruses carrying A at M847 emerged and became the major virus population thereafter, while replicating viruses retained the input A residue at M847 in rZH501-M847-A-infected mice. CONCLUSIONS/SIGNIFICANCE: These data demonstrated that the single nucleotide substitution in the Gn protein substantially affected the RVFV mouse virulence and that a virus population carrying the virulent viral genotype quickly emerged and became the major viral population within a few days in mice that were inoculated with the attenuated virus.


Assuntos
Mutação Puntual , RNA Viral/genética , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/patogenicidade , Animais , Genoma Viral , Camundongos , Polimorfismo de Nucleotídeo Único , Vacinas Atenuadas/efeitos adversos , Virulência/genética
9.
Ann N Y Acad Sci ; 1171 Suppl 1: E75-85, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19751406

RESUMO

Rift Valley fever virus (RVFV), which belongs to the genus Phlebovirus, family Bunyaviridae, is a negative-stranded RNA virus carrying a single-stranded, tripartite RNA genome. RVFV is an important zoonotic pathogen transmitted by mosquitoes and causes large outbreaks among ruminants and humans in Africa and the Arabian Peninsula. Human patients develop an acute febrile illness, followed by a fatal hemorrhagic fever, encephalitis, or ocular diseases. A viral nonstructural protein, NSs, is a major viral virulence factor. Past studies showed that NSs suppresses the transcription of host mRNAs, including interferon-beta mRNAs. Here we demonstrated that the NSs protein induced post-transcriptional downregulation of dsRNA-dependent protein kinase (PKR), to prevent phosphorylation of eIF2alpha and promoted viral translation in infected cells. These two biological activities of the NSs most probably have a synergistic effect in suppressing host innate immune functions and facilitate efficient viral replication in infected mammalian hosts.


Assuntos
RNA Mensageiro/genética , RNA Viral/genética , Febre do Vale de Rift/genética , Vírus da Febre do Vale do Rift/genética , Transcrição Gênica , eIF-2 Quinase/genética , Aborto Espontâneo/virologia , Animais , Culicidae/virologia , Regulação para Baixo , Feminino , Genoma Viral , Humanos , Gravidez , Prenhez , Biossíntese de Proteínas , Vírus de RNA/genética , Febre do Vale de Rift/transmissão , Ruminantes/virologia , Proteínas Virais/genética
10.
PLoS Pathog ; 5(2): e1000287, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19197350

RESUMO

Rift Valley fever virus (RVFV) (genus Phlebovirus, family Bunyaviridae) is a negative-stranded RNA virus with a tripartite genome. RVFV is transmitted by mosquitoes and causes fever and severe hemorrhagic illness among humans, and fever and high rates of abortions in livestock. A nonstructural RVFV NSs protein inhibits the transcription of host mRNAs, including interferon-beta mRNA, and is a major virulence factor. The present study explored a novel function of the RVFV NSs protein by testing the replication of RVFV lacking the NSs gene in the presence of actinomycin D (ActD) or alpha-amanitin, both of which served as a surrogate of the host mRNA synthesis suppression function of the NSs. In the presence of the host-transcriptional inhibitors, the replication of RVFV lacking the NSs protein, but not that carrying NSs, induced double-stranded RNA-dependent protein kinase (PKR)-mediated eukaryotic initiation factor (eIF)2alpha phosphorylation, leading to the suppression of host and viral protein translation. RVFV NSs promoted post-transcriptional downregulation of PKR early in the course of the infection and suppressed the phosphorylated eIF2alpha accumulation. These data suggested that a combination of RVFV replication and NSs-induced host transcriptional suppression induces PKR-mediated eIF2alpha phosphorylation, while the NSs facilitates efficient viral translation by downregulating PKR and inhibiting PKR-mediated eIF2alpha phosphorylation. Thus, the two distinct functions of the NSs, i.e., the suppression of host transcription, including that of type I interferon mRNAs, and the downregulation of PKR, work together to prevent host innate antiviral functions, allowing efficient replication and survival of RVFV in infected mammalian hosts.


Assuntos
Regulação para Baixo , Fator de Iniciação 2 em Eucariotos/metabolismo , Vírus da Febre do Vale do Rift/patogenicidade , Proteínas não Estruturais Virais/fisiologia , Fatores de Virulência/fisiologia , eIF-2 Quinase/metabolismo , Animais , Chlorocebus aethiops , Dactinomicina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Técnicas de Inativação de Genes , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/fisiologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , Fosforilação , RNA Mensageiro/metabolismo , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/crescimento & desenvolvimento , Vírus da Febre do Vale do Rift/fisiologia , Transcrição Gênica/efeitos dos fármacos , Células Vero , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Replicação Viral , eIF-2 Quinase/genética
11.
J Virol ; 81(24): 13335-45, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17913816

RESUMO

Rift Valley fever virus (RVFV) is a member of the genus Phlebovirus within the family Bunyaviridae. It can cause severe epidemics among ruminants and fever, myalgia, a hemorrhagic syndrome, and/or encephalitis in humans. The RVFV M segment encodes the NSm and 78-kDa proteins and two major envelope proteins, Gn and Gc. The biological functions of the NSm and 78-kDa proteins are unknown; both proteins are dispensable for viral replication in cell cultures. To determine the biological functions of the NSm and 78-kDa proteins, we generated the mutant virus arMP-12-del21/384, carrying a large deletion in the pre-Gn region of the M segment. Neither NSm nor the 78-kDa protein was synthesized in arMP-12-del21/384-infected cells. Although arMP-12-del21/384 and its parental virus, arMP-12, showed similar growth kinetics and viral RNA and protein accumulation in infected cells, arMP-12-del21/384-infected cells induced extensive cell death and produced larger plaques than did arMP-12-infected cells. arMP-12-del21/384 replication triggered apoptosis, including the cleavage of caspase-3, the cleavage of its downstream substrate, poly(ADP-ribose) polymerase, and activation of the initiator caspases, caspase-8 and -9, earlier in infection than arMP-12. NSm expression in arMP-12-del21/384-infected cells suppressed the severity of caspase-3 activation. Further, NSm protein expression inhibited the staurosporine-induced activation of caspase-8 and -9, demonstrating that other viral proteins were dispensable for NSm's function in inhibiting apoptosis. RVFV NSm protein is the first identified Phlebovirus protein that has an antiapoptotic function.


Assuntos
Apoptose/efeitos dos fármacos , Vírus da Febre do Vale do Rift/patogenicidade , Proteínas não Estruturais Virais/farmacologia , Animais , Caspases/metabolismo , Linhagem Celular , Chlorocebus aethiops , Ativação Enzimática/efeitos dos fármacos , Humanos , Rim/citologia , Rim/virologia , Macrófagos/virologia , Camundongos , RNA Viral/análise , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/metabolismo , Vírus da Febre do Vale do Rift/fisiologia , Células Vero , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Ensaio de Placa Viral , Replicação Viral
12.
J Virol ; 81(16): 8421-38, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17537856

RESUMO

Rift Valley fever virus (RVFV) (genus Phlebovirus, family Bunyaviridae) has a tripartite negative-strand genome and causes a mosquito-borne disease among humans and livestock in sub-Saharan African and Arabian Peninsula countries. Phlebovirus L, M, and N mRNAs are synthesized from the virus-sense RNA segments, while NSs mRNA is transcribed from the anti-virus-sense S segment. The present study determined the 3' termini of all RVFV mRNAs. The 3' termini of N and NSs mRNAs were mapped within the S-segment intergenic region and were complementary to each other by 30 to 60 nucleotides. The termini of M and L mRNAs were mapped within 122 to 107 nucleotides and 16 to 41 nucleotides, respectively, from the 5' ends of their templates. Viral RNA elements that control phlebovirus transcriptional terminations are largely unknown. Our studies suggested the importance of a pentanucleotide sequence, CGUCG, for N, NSs, and M mRNA transcription terminations. Homopolymeric tracts of C sequences, which were located upstream of the pentanucleotide sequence, promoted N and M mRNA terminations. Likewise, the homopolymeric tracts of G sequences that are found upstream of the pentanucleotide sequence promoted NSs mRNA termination. The L-segment 5'-untranslated region (L-5' UTR) had neither the pentanucleotide sequence nor homopolymeric sequences, yet replacement of the S-segment intergenic region with the L-5' UTR exerted N mRNA termination in an infectious virus. The L-5' UTR contained two 13-nucleotide-long complete complementary sequences, and their sequence complementarities were important for L mRNA termination. A computer-mediated RNA secondary structure analysis further suggested that RNA secondary structures formed by the sections of the two 13-nucleotide-long sequences and by the sequence between them may have a role in L mRNA termination. Our data demonstrated that viral RNA elements that govern L mRNA termination differed from those that regulate mRNA terminations in the M and S segments.


Assuntos
Regiões 3' não Traduzidas/química , Regulação Viral da Expressão Gênica , RNA Viral/química , Vírus da Febre do Vale do Rift/genética , Regiões Terminadoras Genéticas , Regiões 3' não Traduzidas/metabolismo , Sequência de Bases , Genoma Viral/genética , Dados de Sequência Molecular , Conformação de Ácido Nucleico , RNA Viral/metabolismo , Análise de Sequência de RNA , Transcrição Gênica
13.
J Virol ; 80(16): 8274-8, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16873285

RESUMO

Rift Valley fever viruses carrying mutations of the M gene preglycoprotein region, one lacking NSm protein expression, one lacking 78-kDa protein expression, and one lacking expression of both proteins, were compared in cell culture. All of the mutants and their parent virus produced plaques with similar sizes and morphologies in Vero E6 cells and had similar growth kinetics in Vero, C6/36, and MRC5 cells, demonstrating that the NSm and 78-kDa proteins were not needed for the virus to replicate efficiently in cell culture. A competition-propagation assay revealed that the parental virus was slightly more fit than the mutant virus lacking expression of both proteins.


Assuntos
Vírus da Febre do Vale do Rift/fisiologia , Proteínas não Estruturais Virais/fisiologia , Replicação Viral/genética , Animais , Células Cultivadas , Chlorocebus aethiops , Genes Virais/fisiologia , Mutação , Vírus da Febre do Vale do Rift/genética , Células Vero , Proteínas não Estruturais Virais/genética
14.
Mol Cells ; 21(1): 112-20, 2006 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-16511353

RESUMO

It has been suggested that defective interfering (DI) RNA contributes to the persistence of Japanese en-cephalitis virus (JEV). In this study, we characterized molecular and biological aspects of the DI RNA and its relation to viral persistence. We identified a homolo-gous DI virus intimately associated with JEV persis-tence in Vero cells. The production of DI RNA during undiluted serial passages of JEV coincided with the appearance of cells refractory to acute infection with JEV. We also established a Vero cell clone with a per-sistent JEV infection in which the DI RNA co-replicated efficiently at the expense of helper virus. The infectious virus yield of the clone fluctuated dur-ing its growth depending upon the amount of DI RNA accumulated in the previous replication cycle. Identifi-cation of the corresponding negative-sense RNA of the DI RNA indicated that the DI RNA functioned as a replication unit. Most of the DI RNA molecules re-tained their open reading frames despite a large dele-tion, encompassing most of the prM, the entire E, and the 5' half of the NS1 gene. Taken together, these ob-servations suggest that the generation of homologous DI RNA during successive JEV acute infections in Vero cells probably participates actively in persistent JEV infection.


Assuntos
Vírus Defeituosos/genética , Vírus Defeituosos/fisiologia , Vírus da Encefalite Japonesa (Espécie)/genética , Vírus da Encefalite Japonesa (Espécie)/fisiologia , RNA Viral/genética , Interferência Viral/genética , Sequência de Aminoácidos , Animais , Chlorocebus aethiops , Cricetinae , Efeito Citopatogênico Viral , Genoma Viral/genética , Dados de Sequência Molecular , RNA Viral/análise , Inoculações Seriadas , Células Vero , Replicação Viral/genética
15.
J Virol ; 80(6): 2933-40, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16501102

RESUMO

Rift Valley fever virus (RVFV) (genus Phlebovirus, family Bunyaviridae) has a tripartite negative-strand genome, causes a mosquito-borne disease that is endemic in sub-Saharan African countries and that also causes large epidemics among humans and livestock. Furthermore, it is a bioterrorist threat and poses a risk for introduction to other areas. In spite of its danger, neither veterinary nor human vaccines are available. We established a T7 RNA polymerase-driven reverse genetics system to rescue infectious clones of RVFV MP-12 strain entirely from cDNA, the first for any phlebovirus. Expression of viral structural proteins from the protein expression plasmids was not required for virus rescue, whereas NSs protein expression abolished virus rescue. Mutants of MP-12 partially or completely lacking the NSs open reading frame were viable. These NSs deletion mutants replicated efficiently in Vero and 293 cells, but not in MRC-5 cells. In the latter cell line, accumulation of beta interferon mRNA occurred after infection by these NSs deletion mutants, but not after infection by MP-12. The NSs deletion mutants formed larger plaques than MP-12 did in Vero E6 cells and failed to shut off host protein synthesis in Vero cells. An MP-12 mutant carrying a luciferase gene in place of the NSs gene replicated as efficiently as MP-12 did, produced enzymatically active luciferase during replication, and stably retained the luciferase gene after 10 virus passages, representing the first demonstration of foreign gene expression in any bunyavirus. This reverse genetics system can be used to study the molecular virology of RVFV, assess current vaccine candidates, produce new vaccines, and incorporate marker genes into animal vaccines.


Assuntos
DNA Complementar/metabolismo , RNA Polimerases Dirigidas por DNA/metabolismo , Luciferases/metabolismo , Vírus da Febre do Vale do Rift/crescimento & desenvolvimento , Vírus da Febre do Vale do Rift/genética , Proteínas não Estruturais Virais/genética , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , DNA Complementar/genética , Técnicas Genéticas , Humanos , Luciferases/genética , Mutação , Vírus da Febre do Vale do Rift/patogenicidade , Células Vero , Proteínas não Estruturais Virais/metabolismo , Ensaio de Placa Viral , Replicação Viral
16.
J Virol ; 79(18): 12106-11, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16140788

RESUMO

Analysis of purified Rift Valley fever virus (RVFV) particles demonstrated the presence of three negative-sense RNA genomes, plus three anti-viral-sense RNA segments. The virion-associated anti-viral-sense S segment served as a template for the synthesis of NSs mRNA immediately after infection. NSs protein synthesis also occurred early in infection, suggesting that NSs protein produced early in infection probably has biologically significant roles in virus replication and/or survival in the host. Translation inhibitor treatment of mammalian cells infected with viruses belonging to the Bunyaviridae family generally inhibits viral mRNA synthesis. However, RVFV NSs mRNA synthesis, but not N mRNA synthesis, was resistant to puromycin treatment during primary transcription, pointing to the uniqueness of RVFV NSs mRNA synthesis.


Assuntos
RNA Antissenso/genética , RNA Mensageiro/genética , RNA Viral/genética , Vírus da Febre do Vale do Rift/genética , Animais , Chlorocebus aethiops , Inibidores da Síntese de Proteínas/farmacologia , Puromicina/farmacologia , RNA Antissenso/biossíntese , RNA Mensageiro/biossíntese , RNA Viral/biossíntese , Vírus da Febre do Vale do Rift/efeitos dos fármacos , Vírus da Febre do Vale do Rift/fisiologia , Transcrição Gênica , Células Vero , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...