Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Res Sq ; 2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-33619476

RESUMO

One year into the Coronavirus Disease 2019 (COVID-19) pandemic caused by Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), effective treatments are still needed 1-3 . Monoclonal antibodies, given alone or as part of a therapeutic cocktail, have shown promising results in patients, raising the hope that they could play an important role in preventing clinical deterioration in severely ill or in exposed, high risk individuals 4-6 . Here, we evaluated the prophylactic and therapeutic effect of COVA1-18 in vivo , a neutralizing antibody isolated from a convalescent patient 7 and highly potent against the B.1.1.7. isolate 8,9 . In both prophylactic and therapeutic settings, SARS-CoV-2 remained undetectable in the lungs of COVA1-18 treated hACE2 mice. Therapeutic treatment also caused a dramatic reduction in viral loads in the lungs of Syrian hamsters. When administered at 10 mg kg - 1 one day prior to a high dose SARS-CoV-2 challenge in cynomolgus macaques, COVA1-18 had a very strong antiviral activity in the upper respiratory compartments with an estimated reduction in viral infectivity of more than 95%, and prevented lymphopenia and extensive lung lesions. Modelling and experimental findings demonstrate that COVA1-18 has a strong antiviral activity in three different preclinical models and could be a valuable candidate for further clinical evaluation.

2.
J Virol ; 91(2)2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27807226

RESUMO

The retinoic acid-inducible gene 1 (RIG-I) signaling pathway is essential for the recognition of viruses and the initiation of host interferon (IFN)-mediated antiviral responses. Once activated, RIG-I interacts with polyubiquitin chains generated by TRIM25 and binds mitochondrial antiviral signaling protein (MAVS), leading to the production of type I IFN. We now show specific interactions among these key partners in the RLR pathway through the use of bimolecular fluorescence complementation (BiFC) and super-resolution microscopy. Dimers of RIG-I, TRIM25, and MAVS localize into different compartments. Upon activation, we show that TRIM25 is redistributed into cytoplasmic dots associated with stress granules, while RIG-I associates with TRIM25/stress granules and with mitochondrial MAVS. In addition, MAVS competes with TRIM25 for RIG-I binding, and this suggests that upon TRIM25-mediated activation of RIG-I, RIG-I moves away from TRIM25 to interact with MAVS at the mitochondria. For the first time, the distribution of these three proteins was analyzed at the same time in virus-infected cells. We also investigated how specific viral proteins modify some of the protein complexes in the pathway. The protease NS3/4A from hepatitis C virus redistributes the complexes RIG-I/MAVS and MAVS/MAVS but not RIG-I/TRIM25. In contrast, the influenza A virus NS1 protein interacts with RIG-I and TRIM25 in specific areas in the cell cytoplasm and inhibits the formation of TRIM25 homocomplexes but not the formation of RIG-I/TRIM25 heterocomplexes, preventing the formation of RIG-I/MAVS complexes. Thus, we have localized spatially in the cell different complexes formed between RIG-I, TRIM25, and MAVS, in the presence or absence of two viral IFN antagonistic proteins. IMPORTANCE: The first line of defense against viral infections is the innate immune response. Viruses are recognized by pathogen recognition receptors, such as the RIG-I like receptor family, that activate a signaling cascade that induces IFN production. In the present study, we visualized, for the first time in cells, both in overexpression and endogenous levels, complexes formed among key proteins involved in this innate immune signaling pathway. Through different techniques we were able to analyze how these proteins are distributed and reorganized spatially within the cell in order to transmit the signal, leading to an efficient antiviral state. In addition, this work presents a new means by how, when, and where viral proteins can target these pathways and act against the host immune system in order to counteract the activation of the immune response.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína DEAD-box 58/metabolismo , Complexos Multiproteicos/metabolismo , Fatores de Transcrição/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Linhagem Celular , Proteína DEAD-box 58/química , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Espaço Intracelular , Ligação Proteica , Multimerização Proteica , Transporte Proteico , Transdução de Sinais , Fatores de Transcrição/química , Proteínas com Motivo Tripartido/química , Ubiquitina-Proteína Ligases/química , Proteínas não Estruturais Virais/metabolismo
3.
J Theor Biol ; 351: 47-57, 2014 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-24594370

RESUMO

Viral antagonism of host responses is an essential component of virus pathogenicity. The study of the interplay between immune response and viral antagonism is challenging due to the involvement of many processes acting at multiple time scales. Here we develop an ordinary differential equation model to investigate the early, experimentally measured, responses of human monocyte-derived dendritic cells to infection by two H1N1 influenza A viruses of different clinical outcomes: pandemic A/California/4/2009 and seasonal A/New Caledonia/20/1999. Our results reveal how the strength of virus antagonism, and the time scale over which it acts to thwart the innate immune response, differs significantly between the two viruses, as is made clear by their impact on the temporal behavior of a number of measured genes. The model thus sheds light on the mechanisms that underlie the variability of innate immune responses to different H1N1 viruses.


Assuntos
Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/imunologia , Modelos Imunológicos , Células Dendríticas/imunologia , Células Dendríticas/virologia , Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , Imunidade Inata/genética , Imunidade Inata/imunologia , Vírus da Influenza A Subtipo H1N1/classificação , Vírus da Influenza A Subtipo H1N1/patogenicidade , Influenza Humana/genética , Influenza Humana/virologia , Interferon beta/biossíntese , Proteínas não Estruturais Virais/fisiologia
4.
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
5.
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
6.
Arch Virol ; 155(6): 905-14, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20372944

RESUMO

The mechanisms responsible for virulence of influenza viruses in humans remain poorly understood. A prevailing hypothesis is that the highly pathogenic virus isolates cause a severe cytokinemia precipitating acute respiratory distress syndrome and multiple organ dysfunction syndrome. Cynomolgus macaques (Macaca fascicularis) infected with a human highly pathogenic avian influenza (HPAI) H5N1 virus isolate (A/Vietnam/1203/2004) or reassortants of human influenza virus A/Texas/36/91 (H1N1) containing genes from the 1918 pandemic influenza A (H1N1) virus developed severe pneumonia within 24 h postinfection. However, virus spread beyond the lungs was only detected in the H5N1 group, and signs of extrapulmonary tissue reactions, including microglia activation and sustained up-regulation of inflammatory markers, most notably hypoxia inducible factor-1alpha (HIF-1alpha), were largely limited to this group. Extrapulmonary pathology may thus contribute to the morbidities induced by H5N1 viruses.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Virus da Influenza A Subtipo H5N1/patogenicidade , Fígado/patologia , Microglia/imunologia , Infecções por Orthomyxoviridae/fisiopatologia , Animais , Citocinas/metabolismo , Humanos , Macaca fascicularis , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Sistema Respiratório/patologia , Regulação para Cima , Virulência
7.
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
8.
Gene Ther ; 16(6): 796-804, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19242529

RESUMO

Despite the advances in cancer therapies in the past century, malignant melanoma continues to present a significant clinical challenge due to lack of chemotherapeutic response. Systemic therapy with immunostimulatory agents such as interferon and interleukin-2 (IL-2) has shown some promise, though each is associated with significant side effects. Over the past 50 years, oncolytic Newcastle disease virus (NDV) has emerged as an alternative candidate for cancer therapy. The establishment of reverse-genetics systems for the virus has allowed us to further manipulate the virus to enhance its oncolytic activity. Introduction of immunomodulatory molecules, especially IL-2, into the NDV genome was shown to enhance the oncolytic potential of the virus in a murine syngeneic colon carcinoma model. We hypothesize that a recombinant NDV expressing IL-2 would be an effective agent for therapy of malignant melanoma. We show that recombinant NDV possesses a strong cytolytic activity against multiple melanoma cell lines, and is effective in clearing established syngeneic melanoma tumors in mice. Moreover, introduction of murine IL-2 into NDV significantly enhanced its activity against syngeneic melanomas, resulting in increased overall animal survival and generation of antitumor immunity. These findings warrant further investigations of IL-2-expressing NDV as an antimelanoma agent in humans.


Assuntos
Interleucina-2/genética , Melanoma/terapia , Vírus da Doença de Newcastle/genética , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Linfócitos T Citotóxicos/imunologia , Animais , Linhagem Celular Tumoral , Citotoxicidade Imunológica/imunologia , Modelos Animais de Doenças , Intervalo Livre de Doença , Feminino , Humanos , Interleucina-2/metabolismo , Interleucina-2/uso terapêutico , Masculino , Melanoma/imunologia , Camundongos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Recombinação Genética , Transplante Isogênico , Proteínas Virais de Fusão , Replicação Viral
9.
Cancer Gene Ther ; 16(3): 266-78, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18846115

RESUMO

Recombinant oncolytic viruses represent a promising alternative option for the treatment of malignant cancers. We have reported earlier the safety and efficacy of recombinant vesicular stomatitis virus (VSV) vectors in a rat model of hepatocellular carcinoma (HCC). However, the full potential of VSV therapy is limited by a sudden decline in intratumoral virus replication observed early after viral administration, a phenomenon that coincides with an accumulation of inflammatory cells within infected lesions. To overcome the antiviral function of these cells, we present a recombinant virus, rVSV-UL141, which expresses a protein from human cytomegalovirus known to downregulate the natural killer (NK) cell-activating ligand CD155. The modified vector resulted in an inhibition of NK cell recruitment in vitro, as well as decreased intratumoral accumulations of NK and NKT cells in vivo. Administration of rVSV-UL141 through hepatic artery infusion in immune-competent Buffalo rats harboring orthotopic, multi-focal HCC lesions resulted in a one-log elevation of intratumoral virus replication over a control rVSV vector, which translated to enhance tumor necrosis and substantial prolongation of survival. Moreover, these results were achieved in the absence of apparent toxicities. The present study suggests the applicability of this strategy for the development of effective and safe oncolytic agents to treat multi-focal HCC, and potentially a multitude of other cancers, in the future.


Assuntos
Citomegalovirus/genética , Vetores Genéticos/uso terapêutico , Células Matadoras Naturais/imunologia , Neoplasias Hepáticas Experimentais/terapia , Linfócitos do Interstício Tumoral/imunologia , Células T Matadoras Naturais/imunologia , Terapia Viral Oncolítica , Vesiculovirus/fisiologia , Animais , Quimiotaxia de Leucócito , Efeito Citopatogênico Viral , Imunocompetência , Inflamação , Neoplasias Hepáticas Experimentais/imunologia , Masculino , Necrose , Vírus da Doença de Newcastle/genética , Ratos , Ratos Endogâmicos BUF , Organismos Livres de Patógenos Específicos , Vesiculovirus/genética , Replicação Viral
10.
Vaccine ; 26(29-30): 3580-6, 2008 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-18539366

RESUMO

From a stock of A/turkey/Oregon/71-delNS1 (H7N3) virus, which has a 10 nucleotide deletion in the coding region of the NS1 gene, we found that several variants with different sizes of NS genes could be produced by passaging the virus in 10- and 14-day-old embryonating chicken eggs (ECE), but not in 7-day-old ECE or Vero cells. We were able to rescue the reassortant virus that has different sizes of the NS genes and confirmed that those NS genes are genetically stable. By conducting in vivo studies in 2-week-old chickens, we found two plaque purified variants (D-del pc3 and pc4) which can be used as a potential live-attenuated vaccine. The variants were highly attenuated in chickens and did not transmit the virus from infected chickens to uninoculated cage mates. At the same time, the variants induced relatively high antibody titers which conferred good protection against a high dose heterologous virus challenge. Our study indicates that naturally selected NS1 deletion variants might be useful in the development of live-attenuated influenza vaccines in poultry. Furthermore, deletion in the NS1 protein can be potentially useful as a negative marker for a differentiating infected from vaccinated animals (DIVA) approach.


Assuntos
Vírus da Influenza A/genética , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Vírus Reordenados/genética , Vírus Reordenados/imunologia , Proteínas não Estruturais Virais/genética , Animais , Anticorpos Antivirais/sangue , Embrião de Galinha , Galinhas , Chlorocebus aethiops , Influenza Aviária/prevenção & controle , Deleção de Sequência , Vacinas Atenuadas/imunologia , Células Vero
11.
J Virol ; 81(21): 11817-27, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17715226

RESUMO

We are still inadequately prepared for an influenza pandemic due to the lack of a vaccine effective for subtypes to which the majority of the human population has no prior immunity and which could be produced rapidly in sufficient quantities. There is therefore an urgent need to investigate novel vaccination approaches. Using a combination of genomic and traditional tools, this study compares the protective efficacy in macaques of an intrarespiratory live influenza virus vaccine produced by truncating NS1 in the human influenza A/Texas/36/91 (H1N1) virus with that of a conventional vaccine based on formalin-killed whole virus. After homologous challenge, animals in the live-vaccine group had greatly reduced viral replication and pathology in lungs and reduced upper respiratory inflammation. They also had lesser induction of innate immune pathways in lungs and of interferon-sensitive genes in bronchial epithelium. This postchallenge response contrasted with that shortly after vaccination, when more expression of interferon-sensitive genes was observed in bronchial cells from the live-vaccine group. This suggested induction of a strong innate immune response shortly after vaccination with the NS1-truncated virus, followed by greater maturity of the postchallenge immune response, as demonstrated with robust influenza virus-specific CD4+ T-cell proliferation, immunoglobulin G production, and transcriptional induction of T- and B-cell pathways in lung tissue. In conclusion, a single respiratory tract inoculation with an NS1-truncated influenza virus was effective in protecting nonhuman primates from homologous challenge. This protection was achieved in the absence of significant or long-lasting adverse effects and through induction of a robust adaptive immune response.


Assuntos
Sistema Imunitário/virologia , Orthomyxoviridae/genética , Orthomyxoviridae/metabolismo , Proteínas não Estruturais Virais/química , Animais , Biópsia , Sangue/virologia , Brônquios/patologia , Brônquios/virologia , Linfócitos T CD4-Positivos/metabolismo , Epitélio/virologia , Feminino , Regulação Viral da Expressão Gênica , Vírus da Influenza A Subtipo H1N1/metabolismo , Macaca , Masculino , Transcrição Gênica , Proteínas não Estruturais Virais/fisiologia
12.
J Virol ; 80(21): 10813-28, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16928763

RESUMO

Recent outbreaks of avian influenza in humans have stressed the need for an improved nonhuman primate model of influenza pathogenesis. In order to further develop a macaque model, we expanded our previous in vivo genomics experiments with influenza virus-infected macaques by focusing on the innate immune response at day 2 postinoculation and on gene expression in affected lung tissue with viral genetic material present. Finally, we sought to identify signature genes for early infection in whole blood. For these purposes, we infected six pigtailed macaques (Macaca nemestrina) with reconstructed influenza A/Texas/36/91 virus and three control animals with a sham inoculate. We sacrificed one control and two experimental animals at days 2, 4, and 7 postinfection. Lung tissue was harvested for pathology, gene expression profiling, and proteomics. Blood was collected for genomics every other day from each animal until the experimental endpoint. Gross and microscopic pathology, immunohistochemistry, viral gene expression by arrays, and/or quantitative real-time reverse transcription-PCR confirmed successful yet mild infections in all experimental animals. Genomic experiments were performed using macaque-specific oligonucleotide arrays, and high-throughput proteomics revealed the host response to infection at the mRNA and protein levels. Our data showed dramatic differences in gene expression within regions in influenza virus-induced lesions based on the presence or absence of viral mRNA. We also identified genes tightly coregulated in peripheral white blood cells and in lung tissue at day 2 postinoculation. This latter finding opens the possibility of using gene expression arrays on whole blood to detect infection after exposure but prior to onset of symptoms or shedding.


Assuntos
Influenza Humana/genética , Influenza Humana/virologia , Macaca nemestrina/genética , Macaca nemestrina/virologia , Animais , Modelos Animais de Doenças , Expressão Gênica , Perfilação da Expressão Gênica , Genes Virais , Genômica , Humanos , Imunidade Inata , Vírus da Influenza A/genética , Vírus da Influenza A/imunologia , Vírus da Influenza A/patogenicidade , Influenza Humana/imunologia , Influenza Humana/patologia , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Macaca nemestrina/imunologia , Masculino , Modelos Biológicos , Proteômica , Fatores de Tempo
13.
Curr Top Microbiol Immunol ; 283: 249-80, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15298172

RESUMO

Interferons are cytokines secreted in response to viral infections with potent antiviral activity, and they represent a critical component of the innate immune response against viruses. It has now become apparent that many viruses have evolved different mechanisms to counteract the interferon response, allowing their efficient replication and propagation in their hosts. This review discusses how the development of reverse genetics techniques and the increase in our knowledge of the interferon response have led to the discovery of interferon-antagonistic functions of different genes of viruses belonging to the negative-strand RNA virus group. In many cases, these viral genes encode accessory pro- teins that are not required for viral infectivity but are critical for optimal replication and for virulence in the host.


Assuntos
Interferon Tipo I/antagonistas & inibidores , Vírus de RNA/patogenicidade , Proteínas Virais/fisiologia , Animais , Imunidade Inata , Interferon Tipo I/biossíntese , Mutação , Infecções por Vírus de RNA/imunologia , Infecções por Vírus de RNA/virologia , Vírus de RNA/metabolismo , Proteínas não Estruturais Virais/análise , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/fisiologia , Proteínas Virais/análise , Proteínas Virais/química , Virulência , Replicação Viral
14.
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
15.
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
16.
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
17.
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
18.
Cytokine Growth Factor Rev ; 12(2-3): 143-56, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11325598

RESUMO

Response to IFN involves a rapid and direct signal transduction mechanism that quickly reports that presence of extracellular cytokine to the cell nucleus, preserving the specificity inherent in cytokine-receptor interactions to transcriptionally induce expression of a set of genes encoding important antiviral proteins. Establishment of the resulting antiviral state provides a crucial initial line of defense against viral infection. Studies of IFN-deficient cells and animals derived by gene targeting have demonstrated the essential nature of IFN-mediated innate immunity. The long co-evolutionary history of viruses with their hosts has seen the development of a variety of evasive adaptations that allow viruses to circumvent or inactivate host antiviral mechanisms. Further understanding of both host and viral components of this battle may provide important new strategies for vaccine development and creation of novel antiviral compounds.


Assuntos
Antivirais/imunologia , Interferons/imunologia , Vírus/imunologia , Animais , Antivirais/antagonistas & inibidores , Antivirais/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Humanos , Interferons/antagonistas & inibidores , Interferons/biossíntese , Janus Quinase 1 , Camundongos , Proteínas Tirosina Quinases/metabolismo , RNA de Cadeia Dupla/metabolismo , Receptores de Interferon/metabolismo , Fator de Transcrição STAT1 , Transdução de Sinais , Transativadores/genética , Transativadores/metabolismo , Vírus/patogenicidade , eIF-2 Quinase/metabolismo
19.
Vaccine ; 19(15-16): 1960-7, 2001 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-11228366

RESUMO

DNA immunization is a potential vaccination strategy for neonates and infants. We tested the ability of a prototype DNA vaccine against influenza virus to prime lasting immunity when administered to newborn non-human primates. Neonatal DNA vaccination triggered virus-specific and neutralizing antibodies of titers and persistence depending on the vaccine dose. Subsequent exposure to influenza virus, revealed significantly increased recall responses in the baboons vaccinated with DNA during the neonatal stage. The humoral and cellular responses were enhanced in the baboons primed with DNA vaccine as neonates. Thus, neonatal DNA vaccination of non-human primates triggered immune memory that persisted beyond infancy.


Assuntos
Memória Imunológica , Papio/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Fatores Etários , Animais , Animais Recém-Nascidos , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/sangue , Imunidade Celular , Vírus da Influenza A/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Cinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...