Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
J Virol ; 83(12): 6115-24, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19357164

RESUMO

Herpes simplex virus type 1 (HSV-1) acquires its final envelope by budding into cytoplasmic vesicles thought to be derived from trans-Golgi network membranes. This process is facilitated by interactions among the carboxyl termini of viral glycoproteins and tegument proteins. To directly investigate the relative importance of the carboxyl terminus of glycoprotein D (gD) in the presence or absence of gE, a recombinant virus (gDDeltact) was constructed to specify a truncated gD lacking the carboxy-terminal 29 amino acids. Furthermore, two additional recombinant viruses were constructed by mutating from ATG to CTG the initiation codons of gE (gEctg) or both gE and gM (gEctg+gMctg), causing lack of expression of gE or both gE and gM, respectively. A fourth mutant virus was constructed to specify the gEctg+gDDeltact mutations. The replication properties of these viruses were compared to those of a newly constructed recombinant virus unable to express UL20 due to alteration of the two initiation codons of UL20 (UL20ctgctg). All recombinant viruses were constructed by using the double-Red, site-directed mutagenesis system implemented on the HSV-1(F) genome cloned into a bacterial artificial chromosome. The gEctg, gEctg+gMctg, gDDeltact, and gEctg+gDDeltact viruses produced viral plaques on African monkey kidney cells (Vero), as well as other cells, that were on average approximately 30 to 50% smaller than those produced by the wild-type virus HSV-1(F). In contrast, the UL20ctgctg virus produced very small plaques containing three to five cells, as reported previously for the DeltaUL20 virus lacking the entire UL20 gene. Viral replication kinetics of intracellular and extracellular viruses revealed that all recombinant viruses produced viral titers similar to those produced by the wild-type HSV-1(F) virus intracellularly and extracellularly at late times postinfection, with the exception of the UL20ctgctg and DeltaUL20 viruses, which replicated more than two-and-a-half logs less efficiently than HSV-1(F). Electron microscopy confirmed that all viruses, regardless of their different gene mutations, efficiently produced enveloped virions within infected cells, with the exception of the UL20ctgctg and DeltaUL20 viruses, which accumulated high levels of unenveloped virions in the cytoplasm. These results show that the carboxyl terminus of gD and the full-length gE, either alone or in a redundant manner, are not essential in cytoplasmic virion envelopment and egress from infected cells. Similarly, gM and gE do not function alone or in a redundant manner in cytoplasmic envelopment and virion egress, confirming previous findings.


Assuntos
Herpesvirus Humano 1/fisiologia , Proteínas do Envelope Viral/genética , Montagem de Vírus , Animais , Chlorocebus aethiops , Citoplasma/virologia , DNA Viral/genética , Perfilação da Expressão Gênica , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 1/ultraestrutura , Humanos , Mutagênese Sítio-Dirigida , Mutação , Células Vero , Ensaio de Placa Viral , Proteínas Virais/genética
2.
Virus Genes ; 38(2): 204-14, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19130199

RESUMO

West Nile virus (WNV) is a member of the Flaviriridae family, which can cause significant morbidity and mortality in birds, horses, and humans. The WNV-LSU-AR01 strain was isolated from a dead blue jay in Louisiana in 2001. Phylogenetic analysis using 75 full WNV genomes revealed that the LSU-AR01 strain belongs to a distinct subclade among the North American strains. The LSU-AR01 strain differed from the NY-99 prototypic strain by 26 nucleotides causing six amino acid changes. An asparagine-to-lysine change was located immediately proximal to a known CD8(+)T cell epitope in NS4B, while a glutamine-to-lysine change was located within a predicted CD8(+)T cell epitope in NS5. The LSU-AR01 strain caused pronounced neuronal necrosis, perivascular cuffing and gliosis in comparison to the NY-99-infected mice. These results suggest that the previously identified Connecticut strains may contain highly neurovirulent strains such as the LSU-AR01 that have spread in North America.


Assuntos
Genoma Viral , RNA Viral/genética , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/patogenicidade , Animais , Doenças das Aves/virologia , Aves , Análise por Conglomerados , Feminino , Louisiana , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Filogenia , Mutação Puntual , Análise de Sequência de DNA , Virulência , Febre do Nilo Ocidental/patologia , Vírus do Nilo Ocidental/isolamento & purificação
3.
Vaccine ; 27(6): 893-903, 2009 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19070640

RESUMO

Vesicular stomatitis virus (VSV) has been extensively utilized as a viral vector system for the induction of protective immune responses against a variety of pathogens. We constructed recombinant VSVs specifying either the Indiana or Chandipura virus G glycoprotein and expressing the West Nile virus (WNV) envelope (E) glycoprotein. Mice were intranasally vaccinated using a prime (Indiana)-boost (Chandipura) immunization approach and challenged with the virulent WNV-LSU-AR01. Ninety-percent (9 of 10) of the vaccinated mice survived as compared to 10% of the mock-vaccinated mice after WNV lethal challenge. Histopathological examination of brain tissues revealed neuronal necrosis in mock-vaccinated mice but not in vaccinated mice, and vaccinated, but not mock-vaccinated mice developed a strong neutralizing antibody response against WNV. Extensive immunological analysis using polychromatic flow cytometry staining revealed that vaccinated, but not mock-vaccinated mice developed robust cellular immune responses as evidenced by up-regulation of CD4(+) CD154(+) IFNgamma(+) T cells in vaccinated, but not mock-vaccinated mice. Similarly, vaccinated mice developed robust E-glycoprotein-specific CD8(+) T cell immune responses as evidenced by the presence of a high percentage of CD8(+) CD62L(low) IFNgamma(+) cells. In addition, a sizeable population of CD8(+) CD69(+) cells was detected indicating E-specific activation of mature T cells and CD4(+) CD25(+) CD127(low) T regulatory (T reg) cells were down-regulated. These results suggest that VSV-vectored vaccines administered intranasally can efficiently induce protective humoral and cellular immune responses against WNV infections.


Assuntos
Anticorpos Antivirais/sangue , Linfócitos T/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Vesiculovirus/imunologia , Proteínas Estruturais Virais/imunologia , Vacinas contra o Vírus do Nilo Ocidental/imunologia , Animais , Encéfalo/patologia , Cricetinae , Feminino , Interferon gama/biossíntese , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Análise de Sobrevida , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vírus da Estomatite Vesicular Indiana/genética , Vesiculovirus/genética , Proteínas Estruturais Virais/genética , Febre do Nilo Ocidental/prevenção & controle , Vacinas contra o Vírus do Nilo Ocidental/genética
4.
Am J Respir Cell Mol Biol ; 39(2): 198-207, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18367723

RESUMO

We previously described the physicochemical characteristics (particle size, adsorbed polynuclear aromatic hydrocarbons [PAHs], oxygen, and metal content) of butadiene soot (BDS) nanoparticles generated during incomplete combustion of the high-volume industrial petrochemical, 1,3-butadiene. We also demonstrated localization of BDS-delivered PAHs to lipid droplets of murine and human respiratory cells in vitro and up-regulation of biotransformation and oxidative stress responses in these cells. Here, the objective was to determine whether inhalation of BDS nanoparticles promotes up-regulation of Phase I biotransformation enzymes, oxidative stress responses, and inflammation in the lungs of mice. Female Balb/c mice exposed to BDS (5 mg/m(3), 4 h/d, 4 d) were killed immediately or 1 day after final exposure; bronchoalveolar lavage fluid (BALF) was collected from the lungs; total RNA was extracted from one lung and histopathology performed on the other. Histopathology and BALF analysis revealed particle-laden macrophages in airways of BDS-treated mice, accompanied by neutrophilia and epithelial damage. Microarray and qRT-PCR analyses revealed up-regulation of (1) aryl hydrocarbon receptor (AhR)-responsive genes: AhR repressor (Ahrr) and cytochrome P450 IA1 and IB1(Cyp1a1, Cyp1b1); (2) oxidative stress response genes: heme oxygenase 1 (Hmox1), nuclear factor erythroid-derived 2-like 2 (Nfe2l2), NADPH dehydrogenase quinone 1 (Nqo1), and glutathione peroxidase 2 (Gpx2); and (3) pro-inflammatory genes: interleukin-6 (IL-6), C-X-C motif ligand 2 (Cxcl2; analog to human IL-8) and ligand 3 (Cxcl3), and granulocyte chemotactic protein (Cxcl6). Inhalation of PAH-rich, petrochemical combustion-derived nanoparticles causes airway inflammation and induces expression of AhR-associated and oxidative stress response genes, as seen in vitro, plus pro-inflammatory genes.


Assuntos
Poluentes Atmosféricos/toxicidade , Pulmão/efeitos dos fármacos , Nanopartículas/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Pneumonia/metabolismo , Hidrocarbonetos Policíclicos Aromáticos/toxicidade , Fuligem , Animais , Biomarcadores/metabolismo , Biotransformação , Líquido da Lavagem Broncoalveolar/química , Feminino , Perfilação da Expressão Gênica , Exposição por Inalação , Pulmão/metabolismo , Pulmão/patologia , Macrófagos Alveolares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Pneumonia/induzido quimicamente , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia
5.
Am J Respir Cell Mol Biol ; 38(5): 532-40, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18079490

RESUMO

Combustion-generated radicals interact to form polynuclear aromatic hydrocarbons (PAHs), including carcinogens. PAHs aggregate into 20- to 50-nm particles, which extend into branched-chain structures (soots). Incomplete combustion yields black soot particles and black smoke. Many PAHs, including those in soots, fluoresce upon excitation. We have reported that butadiene soot (BDS), generated during combustion of the high-volume petrochemical 1,3-butadiene, serves as a reproducible example of combustion-derived fine and ultrafine particles, with the potential for acute or delayed health effects. Human bronchoepithelial cells (BEAS-2B) display time- and concentration-dependent responses to BDS exposure, culminating in concentration of fluorescent PAHs within discrete cytoplasmic bodies. Here we identify the cytoplasmic compartment(s) in which combustion-derived PAHs concentrate and assess the metabolic responses associated with this compartmentalization. BDS-associated fluorescence colocalized with a red fluorescent cholesterol analog and a transfected plasmid coding for a fluorescent lipid droplet surface protein within BEAS-2B cells. After BDS exposure, murine alveolar macrophages (MH-S) and adipocytes (3T3-L1) also develop fluorescence. These findings, especially within adipocytes, support the accumulation of PAHs within lipid droplets. Microarray data revealed up-regulation of aryl hydrocarbon receptor-induced Phase I biotransformation enzymes and nuclear erythroid-2 related factor 2-mediated oxidative stress responses in BEAS-2B cells. Quantitative RT-PCR results confirmed a time-dependent up-regulation of Phase I biotransformation enzymes (CYP1A1, CYP1B1, and ALDH3A1) in BDS-exposed BEAS-2B and MH-S cells. Thus, respiratory cell lipid droplets concentrate PAHs delivered by combustion-derived ultrafine particles. These PAHs, including several found in BDS and in cigarette smoke, activate xenobiotic metabolism pathways and thereby potentiate their toxicity.


Assuntos
Células Epiteliais/metabolismo , Metabolismo dos Lipídeos , Hidrocarbonetos Policíclicos Aromáticos/metabolismo , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo , Células 3T3-L1 , Animais , Brônquios/citologia , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Butadienos/química , Butadienos/metabolismo , Butadienos/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Camundongos , Microscopia de Fluorescência , Análise de Sequência com Séries de Oligonucleotídeos , Hidrocarbonetos Policíclicos Aromáticos/química , Hidrocarbonetos Policíclicos Aromáticos/toxicidade , Receptores de Hidrocarboneto Arílico/biossíntese , Receptores de Hidrocarboneto Arílico/fisiologia , Mucosa Respiratória/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fuligem/química , Fuligem/metabolismo , Fatores de Tempo
6.
Environ Health Perspect ; 115(12): 1757-66, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18087596

RESUMO

BACKGROUND: In utero environmental tobacco smoke (ETS) exposure exacerbates initial lung responses of adult mice to ovalbumin (OVA), a common allergen in rodent models of allergic asthma. OBJECTIVE: We tested the hypothesis that in utero ETS exposure alters expression of genes (including asthma-related and inflammatory genes) in the lungs of adult mice and that this differential expression is reflected in differential respiratory and immune responses to nontobacco allergens. METHODS: Using Affymetrix Mouse Genome 430 2.0 arrays, we examined gene expression changes in lungs of BALB/c mice exposed to ETS in utero, OVA, or saline aerosol at weeks 7-8, and OVA sensitization and challenge at weeks 11-15. Data sets were filtered by transcript p-value (< or = 0.05), false discovery rate (< or = 0.05), and fold change (> or = 1.5). Differential expression of selected genes was confirmed by polymerase chain reaction (PCR). RESULTS: Genes differentially expressed as a result of in utero ETS exposure are involved in regulation of biological processes (immune response, cell proliferation, apoptosis, cell metabolism) through altered cytoskeleton, adhesion, transcription, and enzyme molecules. A number of genes prominent in lung inflammation were differentially expressed on PCR but did not pass selection criteria for microarray, including arginase (Arg1), chitinases (Chia, Chi3l3, Chi3l4), eotaxins (Ccl11, Ccl24), small proline-rich protein 2a (Sprr2a), and cytokines (Il4, Il6, Il10, Il13, Tnfa) . CONCLUSION: The differential lung gene expression reported here is consistent with previously reported functional changes in lungs of mice exposed in utero to ETS and as adults to the nontobacco allergen OVA.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Efeitos Tardios da Exposição Pré-Natal , Poluição por Fumaça de Tabaco/efeitos adversos , Animais , Asma/genética , Citocinas/genética , Citocinas/metabolismo , Feminino , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Camundongos , Camundongos Endogâmicos BALB C , Análise em Microsséries , Ovalbumina , Gravidez , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA