Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 184
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 87(19): 10435-46, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23903831

RESUMO

Current influenza virus vaccines contain H1N1 (phylogenetic group 1 hemagglutinin), H3N2 (phylogenetic group 2 hemagglutinin), and influenza B virus components. These vaccines induce good protection against closely matched strains by predominantly eliciting antibodies against the membrane distal globular head domain of their respective viral hemagglutinins. This domain, however, undergoes rapid antigenic drift, allowing the virus to escape neutralizing antibody responses. The membrane proximal stalk domain of the hemagglutinin is much more conserved compared to the head domain. In recent years, a growing collection of antibodies that neutralize a broad range of influenza virus strains and subtypes by binding to this domain has been isolated. Here, we demonstrate that a vaccination strategy based on the stalk domain of the H3 hemagglutinin (group 2) induces in mice broadly neutralizing anti-stalk antibodies that are highly cross-reactive to heterologous H3, H10, H14, H15, and H7 (derived from the novel Chinese H7N9 virus) hemagglutinins. Furthermore, we demonstrate that these antibodies confer broad protection against influenza viruses expressing various group 2 hemagglutinins, including an H7 subtype. Through passive transfer experiments, we show that the protection is mediated mainly by neutralizing antibodies against the stalk domain. Our data suggest that, in mice, a vaccine strategy based on the hemagglutinin stalk domain can protect against viruses expressing divergent group 2 hemagglutinins.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vetores Genéticos/administração & dosagem , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A/fisiologia , Vacinas contra Influenza/administração & dosagem , Infecções por Orthomyxoviridae/prevenção & controle , Animais , Especificidade de Anticorpos , Células Cultivadas , Reações Cruzadas , Ensaio de Imunoadsorção Enzimática , Feminino , Imunofluorescência , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Vírus da Influenza A/classificação , Rim/imunologia , Rim/metabolismo , Rim/virologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Filogenia
2.
J Virol ; 87(3): 1290-300, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23175362

RESUMO

The innate immune system is responsible for recognizing invading pathogens and initiating a protective response. In particular, the retinoic acid-inducible gene 1 protein (RIG-I) participates in the recognition of single- and double-stranded RNA viruses. RIG-I activation leads to the production of an appropriate cytokine and chemokine cocktail that stimulates an antiviral state and drives the adaptive immune system toward an efficient and specific response against the ongoing infection. One of the best-characterized natural RIG-I agonists is the defective interfering (DI) RNA produced by Sendai virus strain Cantell. This 546-nucleotide RNA is a well-known activator of the innate immune system and an extremely potent inducer of type I interferon. We designed an in vitro-transcribed RNA that retains the type I interferon stimulatory properties, and the RIG-I affinity of the Sendai virus produced DI RNA both in vitro and in vivo. This in vitro-synthesized RNA is capable of enhancing the production of anti-influenza virus hemagglutinin (HA)-specific IgG after intramuscular or intranasal coadministration with inactivated H1N1 2009 pandemic vaccine. Furthermore, our adjuvant is equally effective at increasing the efficiency of an influenza A/Puerto Rico/8/34 virus inactivated vaccine as a poly(I·C)- or a squalene-based adjuvant. Our in vitro-transcribed DI RNA represents an excellent tool for the study of RIG-I agonists as vaccine adjuvants and a starting point in the development of such a vaccine.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , RNA Helicases DEAD-box/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , RNA Viral/administração & dosagem , Vírus Sendai/genética , Administração Intranasal , Animais , Anticorpos Antivirais/sangue , Proteína DEAD-box 58 , Imunoglobulina G/sangue , Vacinas contra Influenza/administração & dosagem , Injeções Intramusculares , Camundongos , RNA Viral/metabolismo , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia
3.
Equine Vet J ; 41(1): 87-92, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19301588

RESUMO

REASONS FOR PERFORMING STUDY: Three previously described NS1 mutant equine influenza viruses encoding carboxy-terminally truncated NS1 proteins are impaired in their ability to inhibit type I IFN production in vitro and are replication attenuated, and thus are candidates for use as a modified live influenza virus vaccine in the horse. HYPOTHESIS: One or more of these mutant viruses is safe when administered to horses, and recipient horses when challenged with wild-type influenza have reduced physiological and virological correlates of disease. METHODS: Vaccination and challenge studies were done in horses, with measurement of pyrexia, clinical signs, virus shedding and systemic proinflammatory cytokines. RESULTS: Aerosol or intranasal inoculation of horses with the viruses produced no adverse effects. Seronegative horses inoculated with the NS1-73 and NS1-126 viruses, but not the NS1-99 virus, shed detectable virus and generated significant levels of antibodies. Following challenge with wild-type influenza, horses vaccinated with NS1-126 virus did not develop fever (>38.5 degrees C), had significantly fewer clinical signs of illness and significantly reduced quantities of virus excreted for a shorter duration post challenge compared to unvaccinated controls. Mean levels of proinflammatory cytokines IL-1beta and IL-6 were significantly higher in control animals, and were positively correlated with peak viral shedding and pyrexia on Day +2 post challenge. CONCLUSION AND CLINICAL RELEVANCE: These data suggest that the recombinant NS1 viruses are safe and effective as modified live virus vaccines against equine influenza. This type of reverse genetics-based vaccine can be easily updated by exchanging viral surface antigens to combat the problem of antigenic drift in influenza viruses.


Assuntos
Anticorpos Antivirais/sangue , Doenças dos Cavalos/prevenção & controle , Vírus da Influenza A Subtipo H3N8/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/veterinária , Administração Intranasal , Animais , Citocinas/biossíntese , Doenças dos Cavalos/imunologia , Doenças dos Cavalos/virologia , Cavalos , Vacinas contra Influenza/efeitos adversos , Vacinas contra Influenza/genética , Nebulizadores e Vaporizadores/veterinária , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Projetos Piloto , Recombinação Genética , Segurança , Fatores de Tempo , Resultado do Tratamento , Vacinação/veterinária , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/imunologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Eliminação de Partículas Virais
4.
Avian Dis ; 47(3 Suppl): 1047-50, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14575108

RESUMO

Current vaccines to prevent avian influenza rely upon labor-intensive parenteral injection. A more advantageous vaccine would be capable of administration by mass immunization methods such as spray or water vaccination. A recombinant vaccine (rNDV-AIV-H7) was constructed by using a lentogenic paramyxovirus type 1 vector (Newcastle disease virus [NDV] B1 strain) with insertion of the hemagglutinin (HA) gene from avian influenza virus (AIV) A/chicken/NY/13142-5/94 (H7N2). The recombinant virus had stable insertion and expression of the H7 AIV HA gene as evident by detection of HA expression via immunofluorescence in infected Vero cells. The rNDV-AIV-H7 replicated in 9-10 day embryonating chicken eggs and exhibited hemagglutinating activity from both NDV and AI proteins that was inhibited by antisera against both NDV and AIV H7. Groups of 2-week-old white Leghorn chickens were vaccinated with transfectant NDV vector (tNDV), rNDV-AIV-H7, or sterile allantoic fluid and were challenged 2 weeks later with viscerotropic velogenic NDV (vvNDV) or highly pathogenic (HP) AIV. The sham-vaccinated birds were not protected from vvNDV or HP AIV challenge. The transfectant NDV vaccine provided 70% protection for NDV challenge but did not protect against AIV challenge. The rNDV-AIV-H7 vaccine provided partial protection (40%) from vvNDV and HP AIV challenge. The serologic response was examined in chickens that received one or two immunizations of the rNDV-AIV-H7 vaccine. Based on hemagglutination inhibition and enzyme-linked immunosorbent assay (ELISA) tests, chickens that received a vaccine boost seroconverted to AIV H7, but the serologic response was weak in birds that received only one vaccination. This demonstrates the potential for NDV for use as a vaccine vector in expressing AIV proteins.


Assuntos
Vírus da Influenza A/imunologia , Influenza Aviária/imunologia , Doença de Newcastle/imunologia , Vírus da Doença de Newcastle/imunologia , Doenças das Aves Domésticas/imunologia , Vacinas Sintéticas/uso terapêutico , Vacinas Virais/uso terapêutico , Animais , Embrião de Galinha/virologia , Galinhas , Imunização/métodos , Influenza Aviária/prevenção & controle , Doença de Newcastle/prevenção & controle , Doenças das Aves Domésticas/prevenção & controle , Organismos Livres de Patógenos Específicos
5.
Nat Med ; 7(12): 1306-12, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11726970

RESUMO

While searching for alternative reading-frame peptides encoded by influenza A virus that are recognized by CD8+ T cells, we found an abundant immunogenic peptide encoded by the +1 reading frame of PB1. This peptide derives from a novel conserved 87-residue protein, PB1-F2, which has several unusual features compared with other influenza gene products in addition to its mode of translation. These include its absence from some animal (particularly swine) influenza virus isolates, variable expression in individual infected cells, rapid proteasome-dependent degradation and mitochondrial localization. Exposure of cells to a synthetic version of PB1-F2 induces apoptosis, and influenza viruses with targeted mutations that interfere with PB1-F2 expression induce less extensive apoptosis in human monocytic cells than those with intact PB1-F2. We propose that PB1-F2 functions to kill host immune cells responding to influenza virus infection.


Assuntos
Vírus da Influenza A/patogenicidade , Proteínas Mitocondriais/metabolismo , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Apoptose , Sequência de Bases , Sequência Conservada , Cisteína Endopeptidases/metabolismo , Meia-Vida , Células HeLa , Humanos , Proteínas Mitocondriais/genética , Dados de Sequência Molecular , Complexos Multienzimáticos/metabolismo , Oligopeptídeos/genética , Oligopeptídeos/farmacologia , Fases de Leitura Aberta , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacologia , Complexo de Endopeptidases do Proteassoma , Biossíntese de Proteínas , Transporte Proteico , Especificidade da Espécie , Proteínas Virais/genética
6.
J Virol ; 75(23): 11868-73, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11689668

RESUMO

A complete cDNA clone of the Newcastle disease virus (NDV) vaccine strain Hitchner B1 was constructed, and infectious recombinant virus expressing an influenza virus hemagglutinin was generated by reverse genetics. The rescued virus induces a strong humoral antibody response against influenza virus and provides complete protection against a lethal dose of influenza virus challenge in mice, demonstrating the potential of recombinant NDV as a vaccine vector.


Assuntos
Vetores Genéticos , Vírus da Doença de Newcastle/genética , Recombinação Genética , Animais , Anticorpos Antivirais/biossíntese , Linhagem Celular , Embrião de Galinha , Clonagem Molecular , DNA Complementar , Dados de Sequência Molecular , Vírus da Doença de Newcastle/crescimento & desenvolvimento , Vírus da Doença de Newcastle/patogenicidade
7.
Cancer Res ; 61(22): 8188-93, 2001 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11719449

RESUMO

The NS1 protein of influenza virus is a virulence factor that counteracts the PKR-mediated antiviral response by the host. As a consequence, influenza NS1 gene knockout virus delNS1 (an influenza A virus lacking the NS1 open reading frame) fails to replicate in normal cells but produces infectious particles in PKR-deficient cells. Because it is known that oncogenic ras induces an inhibitor of PKR, we addressed the question of whether the delNS1 virus selectively replicates in cells expressing oncogenic ras. We show that upon transfection and expression of oncogenic N-ras, cells become permissive for productive delNS1 virus replication, suggesting that the delNS1 virus has specific oncolytic properties. Viral growth in the oncogenic ras-transfected cells is associated with a reduction of PKR activation during infection. Moreover, treatment of s.c. established N-ras-expressing melanomas in severe combined immunodeficiency mice with the delNS1 virus revealed that this virus has tumor-ablative potentials. The delNS1 virus does not replicate in nonmalignant cell lines such as melanocytes, keratinocytes, or endothelial cells. The apathogenic nature of the delNS1 virus combined with the selective replication properties of this virus in oncogenic ras-expressing cells renders this virus an attractive candidate for the therapy of tumors with an activated ras-signaling pathway.


Assuntos
Genes ras/fisiologia , Vírus da Influenza A/fisiologia , Animais , Transformação Celular Viral/genética , Transformação Celular Viral/fisiologia , Chlorocebus aethiops , Ativação Enzimática , Genes ras/genética , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Masculino , Melanoma/terapia , Melanoma/virologia , Camundongos , Camundongos SCID , Transfecção , Células Tumorais Cultivadas , Células Vero , Proteínas não Estruturais Virais/genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
8.
J Virol ; 75(16): 7375-83, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11462009

RESUMO

Nucleocytoplasmic transport of viral ribonucleoproteins (vRNPs) is an essential aspect of the replication cycle for influenza A, B, and C viruses. These viruses replicate and transcribe their genomes in the nuclei of infected cells. During the late stages of infection, vRNPs must be exported from the nucleus to the cytoplasm prior to transport to viral assembly sites on the cellular plasma membrane. Previously, we demonstrated that the influenza A virus nuclear export protein (NEP, formerly referred to as the NS2 protein) mediates the export of vRNPs. In this report, we suggest that for influenza B and C viruses the nuclear export function is also performed by the orthologous NEP proteins (formerly referred to as the NS2 protein). The influenza virus B and C NEP proteins interact in the yeast two-hybrid assay with a subset of nucleoporins and with the Crm1 nuclear export factor and can functionally replace the effector domain from the human immunodeficiency virus type 1 Rev protein. We established a plasmid transfection system for the generation of virus-like particles (VLPs) in which a functional viral RNA-like chloramphenicol acetyltransferase (CAT) gene is delivered to a new cell. VLPs generated in the absence of the influenza B virus NEP protein were unable to transfer the viral RNA-like CAT gene to a new cell. From these data, we suggest that the nuclear export of the influenza B and C vRNPs are mediated through interaction between NEP proteins and the cellular nucleocytoplasmic export machinery.


Assuntos
Gammainfluenzavirus/fisiologia , Vírus da Influenza B/fisiologia , Proteínas não Estruturais Virais/fisiologia , Transporte Ativo do Núcleo Celular , Animais , Gatos , Linhagem Celular , Cães , Humanos , Replicação Viral
9.
Proc Natl Acad Sci U S A ; 98(5): 2746-51, 2001 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-11226311

RESUMO

The influenza A virus pandemic of 1918-1919 resulted in an estimated 20-40 million deaths worldwide. The hemagglutinin and neuraminidase sequences of the 1918 virus were previously determined. We here report the sequence of the A/Brevig Mission/1/18 (H1N1) virus nonstructural (NS) segment encoding two proteins, NS1 and nuclear export protein. Phylogenetically, these genes appear to be close to the common ancestor of subsequent human and classical swine strain NS genes. Recently, the influenza A virus NS1 protein was shown to be a type I IFN antagonist that plays an important role in viral pathogenesis. By using the recently developed technique of generating influenza A viruses entirely from cloned cDNAs, the hypothesis that the 1918 virus NS1 gene played a role in virulence was tested in a mouse model. In a BSL3+ laboratory, viruses were generated that possessed either the 1918 NS1 gene alone or the entire 1918 NS segment in a background of influenza A/WSN/33 (H1N1), a mouse-adapted virus derived from a human influenza strain first isolated in 1933. These 1918 NS viruses replicated well in tissue culture but were attenuated in mice as compared with the isogenic control viruses. This attenuation in mice may be related to the human origin of the 1918 NS1 gene. These results suggest that interaction of the NS1 protein with host-cell factors plays a significant role in viral pathogenesis.


Assuntos
Surtos de Doenças , Genes Virais , Vírus da Influenza A/genética , Influenza Humana/epidemiologia , Recombinação Genética , Proteínas não Estruturais Virais/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , DNA Complementar , Cães , Humanos , Vírus da Influenza A/patogenicidade , Influenza Humana/virologia , Camundongos , Dados de Sequência Molecular , Fases de Leitura Aberta , Filogenia , Sequências Reguladoras de Ácido Nucleico
10.
J Virol ; 75(4): 1899-908, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11160689

RESUMO

Previous biochemical data identified a host cell fraction, designated RAF-2, which stimulated influenza virus RNA synthesis. A 48-kDa polypeptide (RAF-2p48), a cellular splicing factor belonging to the DEAD-box family of RNA-dependent ATPases previously designated BAT1 (also UAP56), has now been identified as essential for RAF-2 stimulatory activity. Additionally, RAF-2p48 was independently identified as an influenza virus nucleoprotein (NP)-interacting protein, NPI-5, in a yeast two-hybrid screen of a mammalian cDNA library. In vitro, RAF-2p48 interacted with free NP but not with NP bound to RNA, and the RAF-2p48-NP complex was dissociated following addition of free RNA. Furthermore, RAF-2p48 facilitated formation of the NP-RNA complexes that likely serve as templates for the viral RNA polymerase. RAF-2p48 was shown, in both in vitro binding assays and the yeast two-hybrid system, to bind to the amino-terminal region of NP, a domain essential for RNA binding. Together, these observations suggest that RAF-2p48 facilitates NP-RNA interaction, thus leading to enhanced influenza virus RNA synthesis.


Assuntos
Adenosina Trifosfatases/metabolismo , Vírus da Influenza A/metabolismo , Proteínas Nucleares/metabolismo , Nucleoproteínas/metabolismo , RNA Viral/biossíntese , Adenosina Trifosfatases/isolamento & purificação , Regulação Viral da Expressão Gênica , Células HeLa , Humanos , Vírus da Influenza A/genética , Proteínas Nucleares/isolamento & purificação , RNA Helicases/isolamento & purificação , RNA Helicases/metabolismo , Splicing de RNA , RNA Viral/metabolismo , Spliceossomos/metabolismo , Técnicas do Sistema de Duplo-Híbrido
11.
Philos Trans R Soc Lond B Biol Sci ; 356(1416): 1965-73, 2001 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-11779399

RESUMO

The potential threat of another influenza virus pandemic stimulates discussion on how to prepare for such an event. The most reasonable prophylactic approach appears to be the use of effective vaccines. Since influenza and other negative-stranded RNA viruses are amenable to genetic manipulation using transfection by plasmids, it is possible to outline new reverse genetics-based approaches for vaccination against influenza viruses. We suggest three approaches. First, we use a plasmid-only rescue system that allows the rapid generation of high-yield recombinant vaccine strains. Second, we propose developing second-generation live influenza virus vaccines by constructing an attenuated master strain with deletions in the NS1 protein, which acts as an interferon antagonist. Third, we suggest the use of Newcastle disease virus recombinants expressing influenza virus haemagglutinin proteins of pandemic (epizootic) strains as novel vaccine vectors for use in animals and possibly humans.


Assuntos
Vírus da Influenza A/genética , Vacinas contra Influenza/genética , Plasmídeos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Vírus da Doença de Newcastle/genética , Recombinação Genética , Proteínas não Estruturais Virais/genética
12.
J Virol ; 74(24): 11566-73, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11090154

RESUMO

The alpha/beta interferon (IFN-alpha/beta) system represents one of the first lines of defense against virus infections. As a result, most viruses encode IFN antagonistic factors which enhance viral replication in their hosts. We have previously shown that a recombinant influenza A virus lacking the NS1 gene (delNS1) only replicates efficiently in IFN-alpha/beta-deficient systems. Consistent with this observation, we found that infection of tissue culture cells with delNS1 virus, but not with wild-type influenza A virus, induced high levels of mRNA synthesis from IFN-alpha/beta genes, including IFN-beta. It is known that transactivation of the IFN-beta promoter depends on NF-kappaB and several other transcription factors. Interestingly, cells infected with delNS1 virus showed high levels of NF-kappaB activation compared with those infected with wild-type virus. Expression of dominant-negative inhibitors of the NF-kappaB pathway during delNS1 virus infection prevented the transactivation of the IFN-beta promoter, demonstrating a functional link between NF-kappaB activation and IFN-alpha/beta synthesis in delNS1 virus-infected cells. Moreover, expression of the NS1 protein prevented virus- and/or double-stranded RNA (dsRNA)-mediated activation of the NF-kappaB pathway and of IFN-beta synthesis. This inhibitory property of the NS1 protein of influenza A virus was dependent on its ability to bind dsRNA, supporting a model in which binding of NS1 to dsRNA generated during influenza virus infection prevents the activation of the IFN system. NS1-mediated inhibition of the NF-kappaB pathway may thus play a key role in the pathogenesis of influenza A virus.


Assuntos
Vírus da Influenza A , Interferon-alfa/metabolismo , Interferon beta/metabolismo , NF-kappa B/metabolismo , Infecções por Orthomyxoviridae/metabolismo , Proteínas não Estruturais Virais/metabolismo , Animais , Linhagem Celular , Regulação Viral da Expressão Gênica , Interferon-alfa/genética , Interferon beta/genética , Camundongos , NF-kappa B/genética , Infecções por Orthomyxoviridae/genética , Proteínas não Estruturais Virais/genética
13.
Proc Natl Acad Sci U S A ; 97(22): 12289-94, 2000 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-11027311

RESUMO

An assay has been developed that allows the identification of molecules that function as type I IFN antagonists. Using this assay, we have identified an Ebola virus-encoded inhibitor of the type I IFN response, the Ebola virus VP35 protein. The assay relies on the properties of an influenza virus mutant, influenza delNS1 virus, which lacks the NS1 ORF and, therefore, does not produce the NS1 protein. When cells are infected with influenza delNS1 virus, large amounts of type I IFN are produced. As a consequence, influenza delNS1 virus replicates poorly. However, high-efficiency transient transfection of a plasmid encoding a protein that interferes with type I IFN-induced antiviral functions, such as the influenza A virus NS1 protein or the herpes simplex virus protein ICP34.5, rescues growth of influenza delNS1 virus. When plasmids expressing individual Ebola virus proteins were transfected into Madin Darby canine kidney cells, the Ebola virus VP35 protein enhanced influenza delNS1 virus growth more than 100-fold. VP35 subsequently was shown to block double-stranded RNA- and virus-mediated induction of an IFN-stimulated response element reporter gene and to block double-stranded RNA- and virus-mediated induction of the IFN-beta promoter. The Ebola virus VP35 therefore is likely to inhibit induction of type I IFN in Ebola virus-infected cells and may be an important determinant of Ebola virus virulence in vivo.


Assuntos
Interferon Tipo I/antagonistas & inibidores , Nucleoproteínas/fisiologia , Proteínas do Core Viral/fisiologia , Animais , Linhagem Celular , Cães , Humanos , Vírus da Influenza A/genética , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/fisiologia , Proteínas do Nucleocapsídeo , Regiões Promotoras Genéticas , Ribossomos/genética , Proteínas não Estruturais Virais/genética , Replicação Viral
14.
J Virol ; 74(17): 7989-96, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10933707

RESUMO

We present a novel mechanism by which viruses may inhibit the alpha/beta interferon (IFN-alpha/beta) cascade. The double-stranded RNA (dsRNA) binding protein NS1 of influenza virus is shown to prevent the potent antiviral interferon response by inhibiting the activation of interferon regulatory factor 3 (IRF-3), a key regulator of IFN-alpha/beta gene expression. IRF-3 activation and, as a consequence, IFN-beta mRNA induction are inhibited in wild-type (PR8) influenza virus-infected cells but not in cells infected with an isogenic virus lacking the NS1 gene (delNS1 virus). Furthermore, NS1 is shown to be a general inhibitor of the interferon signaling pathway. Inhibition of IRF-3 activation can be achieved by the expression of wild-type NS1 in trans, not only in delNS1 virus-infected cells but also in cells infected with a heterologous RNA virus (Newcastle disease virus). We propose that inhibition of IRF-3 activation by a dsRNA binding protein significantly contributes to the virulence of influenza A viruses and possibly to that of other viruses.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Vírus da Influenza A/fisiologia , Fatores de Transcrição/metabolismo , Ativação Transcricional , Proteínas não Estruturais Virais/metabolismo , Animais , Western Blotting , Linhagem Celular , Embrião de Galinha , Proteínas de Ligação a DNA/antagonistas & inibidores , Humanos , Fator Regulador 3 de Interferon , Interferon beta/metabolismo , Mutação , Vírus da Doença de Newcastle/fisiologia , RNA Mensageiro/metabolismo , Respirovirus/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/antagonistas & inibidores , Transfecção , Proteínas não Estruturais Virais/genética
15.
J Virol ; 74(13): 6203-6, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10846107

RESUMO

The availability of an influenza virus NS1 gene knockout virus (delNS1 virus) allowed us to establish the significance of the biological relationship between the influenza virus NS1 protein and double-stranded-RNA-activated protein kinase (PKR) in the life cycle and pathogenicity of influenza virus. Our results show that the lack of functional PKR permits the delNS1 virus to replicate in otherwise nonpermissive hosts, suggesting that the major function of the influenza virus NS1 protein is to counteract or prevent the PKR-mediated antiviral response.


Assuntos
Vírus da Influenza A/imunologia , Vírus da Influenza A/patogenicidade , Influenza Humana/imunologia , Proteínas não Estruturais Virais/imunologia , Replicação Viral , eIF-2 Quinase/imunologia , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Modelos Animais de Doenças , Humanos , Vírus da Influenza A/crescimento & desenvolvimento , Influenza Humana/genética , Influenza Humana/virologia , Interferons/imunologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição STAT1 , Transativadores/genética , Transativadores/imunologia , Proteínas não Estruturais Virais/genética , eIF-2 Quinase/genética
16.
Proc Natl Acad Sci U S A ; 97(8): 4309-14, 2000 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-10725408

RESUMO

We propose a rational approach to the generation of live viral vaccines: alteration of virally encoded type I IFN antagonists to attenuate virulence while retaining immunogenicity. We have explored this concept by using the influenza virus. Previously we have shown that the NS1 protein of influenza A virus possesses anti-IFN activity. We now present evidence that influenza A and B viruses encoding altered viral NS1 proteins are highly attenuated in the mouse host, yet provide protection from challenge with wild-type viruses.


Assuntos
Vírus da Influenza A/genética , Vírus da Influenza B/genética , Vacinas contra Influenza/genética , Proteínas não Estruturais Virais/genética , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Primers do DNA , Cães , Ensaio de Imunoadsorção Enzimática , Feminino , Vírus da Influenza A/imunologia , Vírus da Influenza B/imunologia , Vacinas contra Influenza/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Mutagênese Sítio-Dirigida , Células Vero , Proteínas não Estruturais Virais/imunologia
17.
FEMS Immunol Med Microbiol ; 27(4): 291-7, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10727884

RESUMO

Outer membrane protein F of Pseudomonas aeruginosa has vaccine efficacy against infection by P. aeruginosa as demonstrated in a variety of animal models. Through the use of synthetic peptides, three surface-exposed epitopes have been identified. These are called peptides 9 (aa 261-274 in the mature F protein, TDAYNQKLSERRAN), 10 (aa 305-318, NATAEGRAINRRVE), and 18 (aa 282-295, NEYGVEGGRVNAVG). Both the peptide 9 and 10 epitopes are protective when administered as a vaccine. In order to develop a vaccine that is suitable for use in humans, including infants with cystic fibrosis, the use of viral vector systems to present the protective epitopes has been investigated. An 11-amino acid portion of epitope 10 (AEGRAINRRVE) was successfully inserted into the antigenic B site of the hemagglutinin on the surface of influenza virus. This chimeric influenza virus protects against challenge with P. aeruginosa in the mouse model of chronic pulmonary infection. Attempts to derive a chimeric influenza virus carrying epitope 9 have been unsuccessful. A chimeric plant virus, cowpea mosaic virus (CPMV), with epitopes 18 and 10 expressed in tandem on the large coat protein subunit (CPMV-PAE5) was found to elicit antibodies that reacted exclusively with the 10 epitope and not with epitope 18. Use of this chimeric virus as a vaccine afforded protection against challenge with P. aeruginosa in the mouse model of chronic pulmonary infection. Chimeric CPMVs with a single peptide containing epitopes 9 and 18 expressed on either of the coat proteins are in the process of being evaluated. Epitope 9 was successfully expressed on the coat protein of tobacco mosaic virus (TMV), and this chimeric virus is protective when used as a vaccine in the mouse model of chronic pulmonary infection. However, initial attempts to express epitope 10 on the coat protein of TMV have been unsuccessful. Efforts are continuing to construct chimeric viruses that express both the 9 and 10 epitopes in the same virus vector system. Ideally, the use of a vaccine containing two epitopes of protein F is desirable in order to greatly reduce the likelihood of selecting a variant of P. aeruginosa that escapes protective antibodies in immunized humans via a mutation in a single epitope within protein F. When the chimeric influenza virus containing epitope 10 and the chimeric TMV containing epitope 9 were given together as a combined vaccine, the immunized mice produced antibodies directed toward both epitopes 9 and 10. The combined vaccine afforded protection against challenge with P. aeruginosa in the chronic pulmonary infection model at approximately the same level of efficacy as provided by the individual chimeric virus vaccines. These results prove in principle that a combined chimeric viral vaccine presenting both epitopes 9 and 10 of protein F has vaccine potential warranting continued development into a vaccine for use in humans.


Assuntos
Vacinas Bacterianas/imunologia , Vírus da Influenza A/genética , Pneumopatias/prevenção & controle , Vírus de Plantas/genética , Porinas/imunologia , Infecções por Pseudomonas/prevenção & controle , Pseudomonas aeruginosa/imunologia , Animais , Anticorpos Antibacterianos/sangue , Vacinas Bacterianas/administração & dosagem , Comovirus/genética , Comovirus/metabolismo , Ensaio de Imunoadsorção Enzimática , Epitopos/genética , Epitopos/imunologia , Epitopos/metabolismo , Vírus da Influenza A/metabolismo , Pulmão/microbiologia , Pneumopatias/microbiologia , Camundongos , Vírus de Plantas/metabolismo , Porinas/química , Porinas/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Vírus do Mosaico do Tabaco/genética , Vírus do Mosaico do Tabaco/metabolismo , Vacinação , Vacinas Combinadas/administração & dosagem , Vacinas Combinadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
18.
J Gen Virol ; 81(Pt 3): 737-42, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10675411

RESUMO

We have previously obtained four transfectant influenza A viruses containing neuraminidase (NA) genes with mutated base pairs in the conserved double-stranded RNA region of the viral promoter by using a ribonucleoprotein transfection system. Two mutant viruses (D2 and D1/2) which share a C-G-->A-U mutation at positions 11 and 12 of the 3' and 5' ends, respectively, of the NA gene, showed an approximate 10-fold reduction of NA-specific mRNA and protein levels (Fodor et al., Journal of Virology 72, 6283-6290, 1998). These viruses have now allowed us to determine the effects of decreased NA levels on virus pathogenicity. Both D2 and D1/2 viruses were highly attenuated in mice, and their replication in mouse lungs was highly compromised as compared with wild-type influenza A/WSN/33 virus. The results highlight the importance of the level of NA activity in the biological cycle and virulence of influenza viruses. Importantly, mice immunized by a single intranasal administration of 10(3) infectious units of D2 or D1/2 viruses were protected against challenge with a lethal dose of wild-type influenza virus. Attenuation of influenza viruses by mutations resulting in the decreased expression of a viral protein represents a novel strategy which could be considered for the generation of live attenuated influenza virus vaccines.


Assuntos
Vírus da Influenza A/enzimologia , Vírus da Influenza A/patogenicidade , Neuraminidase/metabolismo , Animais , Linhagem Celular , DNA Viral/genética , Feminino , Imunização , Vírus da Influenza A/genética , Vacinas contra Influenza/genética , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Neuraminidase/genética , Infecções por Orthomyxoviridae/prevenção & controle , Fenótipo , Transfecção , Vacinas Atenuadas/genética , Virulência/genética , Virulência/fisiologia , Replicação Viral/genética , Replicação Viral/fisiologia
19.
Cancer Res ; 60(24): 6972-6, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11156398

RESUMO

We are investigating the potential use of influenza virus vectors expressing selected tumor-associated antigens (TAAs) as therapeutic agents in anticancer strategies. Previously, we have shown that recombinant influenza viruses expressing a model TAA mediated the regression of established pulmonary metastases in mice through the induction of cytotoxic T-cell responses (N. P. Restifo et al., Virology, 249: 89-97, 1998). We have now expanded these observations in the mouse model using survival as the end point of the assay. Animals with a high tumor burden showed extended survival times when treated with a recombinant influenza virus expressing a TAA, but they finally succumbed to death. Death was associated with the presence of a small number of large tumors in lungs. Interestingly, these tumors were found to express undetectable levels of the TAAs because of a down-regulation in the TAA-specific mRNA levels. On the other hand, mice with five times lower tumor burden showed complete tumor regression and survival for >6 six months when treated with the recombinant virus. These animals showed protection against a tumor challenge 6 months after treatment. Our results suggest that recombinant influenza viruses may be useful as therapeutic agents for the prevention and treatment of cancers with known TAAs.


Assuntos
Vetores Genéticos , Neoplasias/terapia , Orthomyxoviridae/genética , Animais , Biomarcadores Tumorais/metabolismo , Bovinos , Linhagem Celular , Células Cultivadas , Regulação para Baixo , Feminino , Humanos , Neoplasias Pulmonares/terapia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas , beta-Galactosidase/metabolismo
20.
J Virol ; 73(11): 9679-82, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10516084

RESUMO

We have rescued influenza A virus by transfection of 12 plasmids into Vero cells. The eight individual negative-sense genomic viral RNAs were transcribed from plasmids containing human RNA polymerase I promoter and hepatitis delta virus ribozyme sequences. The three influenza virus polymerase proteins and the nucleoprotein were expressed from protein expression plasmids. This plasmid-based reverse genetics technique facilitates the generation of recombinant influenza viruses containing specific mutations in their genes.


Assuntos
DNA Recombinante/genética , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Animais , Linhagem Celular , Técnicas Genéticas , Humanos , Plasmídeos/genética , RNA Viral/metabolismo , Transcrição Gênica , Transfecção , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA