Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Vaccine Immunol ; 19(11): 1792-7, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22956656

RESUMO

Avian influenza virus causes outbreaks in domestic and wild birds around the world, and sporadic human infections have been reported. A DNA vaccine encoding hemagglutinin (HA) protein from the A/Indonesia/5/05 (H5N1) strain was initially tested in two randomized phase I clinical studies. Vaccine Research Center study 304 (VRC 304) was a double-blinded study with 45 subjects randomized to placebo, 1 mg of vaccine, or 4 mg of vaccine treatment groups (n = 15/group) by intramuscular (i.m.) Biojector injection. VRC 305 was an open-label study to evaluate route, with 44 subjects randomized to intradermal (i.d.) injections of 0.5 mg by needle/syringe or by Biojector or 1 mg delivered as two 0.5-mg Biojector injections in the same deltoid or as 0.5 mg in each deltoid (n = 11/group). Injections were administered at weeks 0, 4, and 8 in both studies. Antibody responses to H5 were assessed by hemagglutination inhibition (HAI) assay, enzyme-linked immunosorbent assay (ELISA), and neutralization assay, and the H5 T cell responses were assessed by enzyme-linked immunospot and intracellular cytokine staining assays. There were no vaccine-related serious adverse events, and the vaccine was well tolerated in all groups. At 1 mg, i.d. vaccination compared to i.m. vaccination induced a greater frequency and magnitude of response by ELISA, but there were no significant differences in the frequency or magnitude of response between the i.d. and i.m. routes in the HAI or neutralization assays. T cell responses were more common in subjects who received the 1- or 4-mg dose i.m. These studies demonstrated that the DNA vaccine encoding H5 is safe and immunogenic and served to define the proper dose and route for further studies. The i.d. injection route did not offer a significant advantage over the i.m. route, and no difference was detected by delivery to one site versus splitting the dose between two sites for i.d. vaccine administration. The 4-mg dose (i.m) was further investigated in prime-boost regimens.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Influenza Humana/prevenção & controle , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Adulto , Anticorpos Antivirais/sangue , Citocinas/metabolismo , Método Duplo-Cego , Ensaio de Imunoadsorção Enzimática , ELISPOT , Feminino , Testes de Inibição da Hemaglutinação , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Vacinas contra Influenza/genética , Injeções Intradérmicas , Injeções Intramusculares , Masculino , Pessoa de Meia-Idade , Testes de Neutralização , Placebos/administração & dosagem , Linfócitos T/imunologia , Vacinas de DNA/genética , Adulto Jovem
2.
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
3.
Poult Sci ; 88(4): 872-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19276438

RESUMO

Influenza viruses with novel hemagglutinin and 1 or more accompanying genes derived from avian influenza viruses sporadically emerge in humans and have the potential to result in a pandemic if the virus causes disease and spreads efficiently in a population that lacks immunity to the novel hemagglutinin. Since 1997, multiple avian influenza virus subtypes have been transmitted directly from domestic poultry to humans and have caused a spectrum of human disease, from asymptomatic to severe and fatal. To assess the pandemic risk that avian influenza viruses pose, we have used multiple strategies to better understand the capacity of avian viruses to infect, cause disease, and transmit among mammals, including humans. Seroepidemiologic studies that evaluate the frequency and risk of human infection with avian influenza viruses in populations with exposure to domestic or wild birds can provide a better understanding of the pandemic potential of avian influenza subtypes. Investigations conducted in Hong Kong following the first H5N1 outbreak in humans in 1997 determined that exposure to poultry in live bird markets was a key risk factor for human disease. Among poultry workers, butchering and exposure to sick poultry were risk factors for antibody to H5 virus, which provided evidence for infection. A second risk assessment tool, the ferret, can be used to evaluate the level of virulence and potential for host-to-host transmission of avian influenza viruses in this naturally susceptible host. Avian viruses isolated from humans exhibit a level of virulence and transmissibility in ferrets that generally reflects that seen in humans. The ferret model thus provides a means to monitor emerging avian influenza viruses for pandemic risk, as well as to evaluate laboratory-generated reassortants and mutants to better understand the molecular basis of influenza virus transmissibility. Taken together, such studies provide valuable information with which we can assess the public health risk of avian influenza viruses.


Assuntos
Influenza Aviária/prevenção & controle , Influenza Humana/prevenção & controle , Saúde Pública , Animais , Aves , Surtos de Doenças , Furões , Saúde Global , Humanos , Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A , Influenza Humana/transmissão , Medição de Risco , Estudos Soroepidemiológicos
4.
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
5.
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
6.
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
7.
Avian Dis ; 47(3 Suppl): 951-5, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14575093

RESUMO

The introduction of an influenza A virus possessing a novel hemagglutinin (HA) into an immunologically naive human population has the potential to cause severe disease and death. Such was the case in 1997 in Hong Kong, where H5N1 influenza was transmitted to humans from infected poultry. Because H5N1 viruses are still isolated from domestic poultry in southern China, there needs to be continued surveillance of poultry and characterization of virus subtypes and variants. This study provides molecular characterization and evaluation of pathogenesis of a recent H5N1 virus isolated from duck meat that had been imported to South Korea from China. The HA gene of A/Duck/Anyang/AVL-1/01 (H5N1) isolate was found to be closely related to the Hong Kong/97 H5N1 viruses. This virus also contained multiple basic amino acids adjacent to the cleavage site between HA1 and HA2, characteristic of high-pathogenicity avian influenza viruses (HPAI). The pathogenesis of this virus was characterized in chickens, ducks, and mice. The DK/Anyang/AVL-1/01 isolate replicated well in all species and resulted in 100% and 22% lethality for chickens and mice, respectively. No clinical signs of disease were observed in DK/Anyang/AVL-1/01-inoculated ducks, but high titers of infectious virus could be detected in multiple tissues and oropharyngeal swabs. The presence of an H5N1 influenza virus in ducks bearing a HA gene that is highly similar to those of the pathogenic 1997 human/poultry H5N1 viruses raises the possibility of reintroduction of HPAI to chickens and humans.


Assuntos
Patos/virologia , Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A/patogenicidade , Carne/virologia , Animais , Galinhas , China , Glicoproteínas de Hemaglutininação de Vírus da Influenza/isolamento & purificação , Vírus da Influenza A/classificação , Vírus da Influenza A/isolamento & purificação , Influenza Aviária/transmissão , Coreia (Geográfico) , Masculino , Camundongos , Camundongos Endogâmicos BALB C
8.
Avian Dis ; 47(3 Suppl): 968-71, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14575095

RESUMO

Apoptosis is essential in many physiological processes including wound healing and development of the immune response. Apoptosis also plays an important role in the pathogenesis of many infectious diseases including those caused by viruses. Influenza viruses induce apoptosis in cells that are permissive for viral replication and cells that do not support viral replication. The cellular pathways involved in influenza virus induced apoptosis are currently ill defined. Previous studies suggest that influenza virus infection increased the expression of the Fas antigen in HeLa cells, and that Fas antigen is partially involved in apoptosis. In these studies we examined the cellular pathways involved in avian influenza virus induced apoptosis in two cell lines that support productive viral replication: Madin-Darby canine kidney cells (MDCK) and mink lung epithelial (Mv1Lu) cells.


Assuntos
Vírus da Influenza A/patogenicidade , Alantoide/virologia , Animais , Apoptose , Linhagem Celular , Embrião de Galinha/virologia , Fragmentação do DNA , Cães , Vison , Necrose
9.
J Virol ; 75(11): 5141-50, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11333895

RESUMO

Influenza vaccines that induce greater cross-reactive or heterosubtypic immunity (Het-I) may overcome limitations in vaccine efficacy imposed by the antigenic variability of influenza A viruses. We have compared mucosal versus traditional parenteral administration of inactivated influenza vaccine for the ability to induce Het-I in BALB/c mice and evaluated a modified Escherichia coli heat-labile enterotoxin adjuvant, LT(R192G), for augmentation of Het-I. Mice that received three intranasal (i.n.) immunizations of H3N2 vaccine in the presence of LT(R192G) were completely protected against lethal challenge with a highly pathogenic human H5N1 virus and had nasal and lung viral titers that were at least 2,500-fold lower than those of control mice receiving LT(R192G) alone. In contrast, mice that received three vaccinations of H3N2 vaccine subcutaneously in the presence or absence of LT(R192G) or incomplete Freund's adjuvant were not protected against lethal challenge and had no significant reductions in tissue virus titers observed on day 5 post-H5N1 virus challenge. Mice that were i.n. administered H3N2 vaccine alone, without LT(R192G), displayed partial protection against heterosubtypic challenge. The immune mediators of Het-I were investigated. The functional role of B and CD8+ T cells in Het-I were evaluated by using gene-targeted B-cell (IgH-6(-/-))- or beta2-microglobulin (beta2m(-/-))-deficient mice, respectively. beta2m(-/-) but not IgH-6(-/-) vaccinated mice were protected by Het-I and survived a lethal infection with H5N1, suggesting that B cells, but not CD8+ T cells, were vital for protection of mice against heterosubtypic challenge. Nevertheless, CD8+ T cells contributed to viral clearance in the lungs and brain tissues of heterotypically immune mice. Mucosal but not parenteral vaccination induced subtype cross-reactive lung immunoglobulin G (IgG), IgA, and serum IgG anti-hemagglutinin antibodies, suggesting the presence of a common cross-reactive epitope in the hemagglutinins of H3 and H5. These results suggest a strategy of mucosal vaccination that stimulates cross-protection against multiple influenza virus subtypes, including viruses with pandemic potential.


Assuntos
Linfócitos B/imunologia , Proteínas de Escherichia coli , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Influenza Aviária/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Administração Cutânea , Administração Intranasal , Animais , Anticorpos Antivirais/análise , Anticorpos Antivirais/sangue , Toxinas Bacterianas/administração & dosagem , Aves , Linfócitos T CD8-Positivos/imunologia , Reações Cruzadas , Enterotoxinas/administração & dosagem , Escherichia coli/imunologia , Feminino , Adjuvante de Freund/administração & dosagem , Hemaglutininas Virais/imunologia , Humanos , Imunoglobulina A/análise , Imunoglobulina A/sangue , Imunoglobulina G/análise , Imunoglobulina G/sangue , Virus da Influenza A Subtipo H5N1/isolamento & purificação , Influenza Aviária/imunologia , Influenza Aviária/virologia , Pulmão/imunologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade da Espécie , Vacinas de Produtos Inativados/imunologia
10.
J Virol ; 75(10): 4896-901, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11312361

RESUMO

Avian influenza A H9N2 viruses are widespread among domestic poultry and were recently isolated from humans with respiratory illness in China. Two antigenically and genetically distinct groups of H9N2 viruses (G1 and G9) are prevalent in China. To evaluate a strategy for vaccination, we compared G1 and G9 viruses for their relative immunogenicity and cross-protective efficacy. Infection of BALB/c mice with representative viruses of either group protected against subsequent challenge with the homologous or heterologous H9N2 virus in the absence of detectable cross-reactive serum hemagglutination inhibition antibody. Mice injected intramuscularly with inactivated G1 whole virus vaccine were completely protected from challenge with either H9N2 virus. In contrast, mice administered inactivated G9 vaccine were only partially protected against heterologous challenge with the G1 virus. These results have implications for the development of human vaccines against H9N2 viruses, a priority for pandemic preparedness.


Assuntos
Vírus da Influenza A Subtipo H9N2 , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Animais , Reações Cruzadas , Modelos Animais de Doenças , Feminino , Humanos , Vírus da Influenza A/fisiologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Vacinação , Vacinas de Produtos Inativados , Replicação Viral
11.
J Virol ; 74(22): 10807-10, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11044127

RESUMO

Highly pathogenic avian influenza A H5N1 viruses caused an outbreak of human respiratory illness in Hong Kong. Of 15 human H5N1 isolates characterized, nine displayed a high-, five a low-, and one an intermediate-pathogenicity phenotype in the BALB/c mouse model. Sequence analysis determined that five specific amino acids in four proteins correlated with pathogenicity in mice. Alone or in combination, these specific residues are the likely determinants of virulence of human H5N1 influenza viruses in this model.


Assuntos
Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A/genética , Vírus da Influenza A/patogenicidade , Influenza Humana/virologia , Adolescente , Adulto , Animais , Pré-Escolar , Modelos Animais de Doenças , Feminino , Humanos , Lactente , Vírus da Influenza A/classificação , Influenza Humana/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Dados de Sequência Molecular , Fenótipo , Análise de Sequência de DNA , Proteínas Virais/genética , Virulência
12.
J Virol ; 74(13): 6105-16, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10846094

RESUMO

Previously, we observed that several virulent influenza A (H5N1) viruses which caused severe or fatal disease in humans were also lethal in BALB/c mice following dissemination of the virus to solid organs, including the brain. In contrast, one particular human H5N1 virus was nonlethal in mice and showed no evidence of systemic spread. To compare H5N1 viruses of varying pathogenicity for their ability to alter the mammalian immune system, mice were infected with either influenza A/Hong Kong/483/97 (HK/483) (lethal) or A/Hong Kong/486/97 (HK/486) (nonlethal) virus and monitored for lymphocyte depletion in the blood, lungs, and lymphoid tissue. Intranasal infection with HK/483 resulted in a significant decrease in the total number of circulating leukocytes evident as early as day 2 postinfection. Differential blood counts demonstrated up to an 80% drop in lymphocytes by day 4 postinfection. In contrast, nonlethal HK/486-infected mice displayed only a transient drop of lymphocytes during the infectious period. Analysis of lung and lymphoid tissue from HK/483-infected mice demonstrated a reduction in the number of CD4(+) and CD8(+) T cells and reduced synthesis of the cytokines interleukin-1beta and gamma interferon and the chemokine macrophage inflammatory protein compared with HK/486-infected mice. In contrast, the cytokine and chemokine levels were increased in the brains of mice infected with HK/483 but not HK/486. Evidence of apoptosis in the spleen and lung of HK/483-infected mice was detected in situ, suggesting a mechanism for lymphocyte destruction. These results suggest that destructive effects on the immune system may be one factor that contributes to the pathogenesis of H5N1 viruses in mammalian hosts.


Assuntos
Citocinas/biossíntese , Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A/imunologia , Influenza Humana/imunologia , Animais , Apoptose , Modelos Animais de Doenças , Feminino , Humanos , Vírus da Influenza A/patogenicidade , Influenza Humana/mortalidade , Pulmão/citologia , Depleção Linfocítica , Linfopenia/virologia , Camundongos , Camundongos Endogâmicos BALB C , Baço/citologia , Virulência
13.
Biomed Pharmacother ; 54(4): 178-87, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10872716

RESUMO

In 1997 in Hong Kong, 18 human cases of respiratory illness were caused by an avian influenza A H5N1 virus. Although avian influenza viruses had not previously been known to cause respiratory illness in humans, the H5N1 viruses caused severe illness and death, primarily in individuals aged > 12 years. The introduction of H5N1 viruses into humans raised concerns about the potential of these viruses to cause a pandemic. We have used the BALB/c mouse to better understand the pathogenesis of and immunity to the H5N1 viruses in a mammalian model. Previously, we demonstrated that H5N1 viruses isolated from humans replicated efficiently in the lungs of mice without prior adaptation to this host. Two general phenotypes of pathogenicity of H5N1 viruses, based on high and low lethality for mice, were observed. We now demonstrate that in addition to a lethal outcome, H5N1 viruses with a high pathogenicity phenotype exhibit additional features that include rapid and uncontrolled replication in the lungs of infected mice, dissemination and replication of the virus in other organs, and depletion of peripheral blood leukocytes. The BALB/c mouse model was also used to better understand the parameters of protective immunity to the H5N1 viruses. Prior infection with H5N1 viruses of low pathogenicity or an antigenically related non-pathogenic H5N3 virus protected mice from death by infection with a highly pathogenic HK/483 virus. Serum hemagglutination-inhibition antibody titers of 40 or greater were associated with protection of mice from death. Immunization of mice with baculovirus-expressed recombinant H5 hemagglutinin protein or a previously defined HS-specific synthetic peptide induced MHC class II restricted CTL activity. Mice that had CTL activity but no serum hemagglutination-inhibition antibody were not protected from a lethal challenge with H5N1 virus. These results suggest that antibody is required for protection of mice against lethal challenge with H5N1 viruses of the high pathogenicity phenotype.


Assuntos
Vírus da Influenza A/imunologia , Vírus da Influenza A/patogenicidade , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/análise , Feminino , Imunização , Vacinas contra Influenza/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/virologia , Linfócitos T Citotóxicos/imunologia , Replicação Viral
14.
Invest Ophthalmol Vis Sci ; 41(6): 1402-9, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10798656

RESUMO

PURPOSE: To investigate the role macrophages play in controlling herpes simplex virus (HSV)-1 replication after infection of the murine cornea. METHODS: Macrophage depletion in selected tissues before or after virus infection was achieved by repeated subconjunctival (SCJ) and/or intravenous (IV) injection of liposomes containing dichloromethylene diphosphonate (L-Cl2MDP). Controls received liposomes containing phosphate-buffered saline (L-PBS). The efficiency of depletion was evaluated by histologic examination. Virus content in infected tissues was determined by standard plaque assay. Delayed-type hypersensitivity (DTH) responsiveness was assessed using the ear-swelling assay. Antibody isotype responses to virus antigens and cytokine production were monitored by enzyme-linked immunosorbent assay. RESULTS: Balb/c mice given SCJ injection of L-Cl2MDP 4 and 2 days before HSV-1 corneal infection were found to have ocular virus titers as much as 10(5)-fold higher than that seen in the L-PBS-treated controls 8 days after infection. When L-Cl2MDP treatment was delayed until 2 and 4 days after infection, virus titers in the eye were analogous to those in the control animals. Subconjunctival and submandibular lymph node macrophages in mice given local (SCJ) L-Cl2MDP pretreatment were profoundly reduced, whereas the number of corneal Langerhans' cells and lymph node dendritic cells remained unchanged. Local L-Cl2MDP pretreatment was associated with significantly reduced DTH responsiveness to HSV-1 antigen, and an alteration in selected antibody isotype production. Depletion of macrophages in the subconjunctival tissue before corneal infection was not accompanied by enhanced virus growth at early times (2 or 4 days) after infection. CONCLUSIONS: Macrophages play an important role in restricting HSV-1 growth after corneal infection. These cells appear to be required for the development of an acquired immune response, presumably by functioning in antigen processing and presentation. The hypothesis that macrophages are major participants in innate immunity to HSV-1 corneal infection was not supported.


Assuntos
Córnea/virologia , Herpesvirus Humano 1/crescimento & desenvolvimento , Ceratite Herpética/virologia , Macrófagos/fisiologia , Replicação Viral , Fosfatase Ácida/metabolismo , Animais , Anticorpos Antivirais/biossíntese , Ácido Clodrônico/farmacologia , Córnea/enzimologia , Córnea/patologia , Citocinas/biossíntese , Células Dendríticas/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Feminino , Hipersensibilidade Tardia/imunologia , Técnicas Imunoenzimáticas , Ceratite Herpética/enzimologia , Ceratite Herpética/imunologia , Ceratite Herpética/patologia , Células de Langerhans/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Replicação Viral/efeitos dos fármacos
15.
J Virol ; 73(7): 5903-11, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10364342

RESUMO

During 1997 in Hong Kong, 18 human cases of respiratory illness, including 6 fatalities, were caused by highly pathogenic avian influenza A (H5N1) viruses. Since H5 viruses had previously been isolated only from avian species, the outbreak raised questions about the ability of these viruses to cause severe disease and death in humans. To better understand the pathogenesis and immunity to these viruses, we have used the BALB/c mouse model. Four H5N1 viruses replicated equally well in the lungs of mice without prior adaptation but differed in lethality for mice. H5N1 viruses that were highly lethal for mice were detected in multiple organs, including the brain. This is the first demonstration of an influenza A virus that replicates systemically in a mammalian species and is neurotropic without prior adaptation. The mouse model was also used to evaluate a strategy of vaccination against the highly pathogenic avian H5N1 viruses, using an inactivated vaccine prepared from nonpathogenic A/Duck/Singapore-Q/F119-3/97 (H5N3) virus that was antigenically related to the human H5N1 viruses. Mice administered vaccine intramuscularly, with or without alum, were completely protected from lethal challenge with H5N1 virus. Protection from infection was also observed in 70% of animals administered vaccine alone and 100% of mice administered vaccine with alum. The protective effect of vaccination correlated with the level of virus-specific serum antibody. These results suggests a strategy of vaccine preparedness for rapid intervention in future influenza pandemics that uses antigenically related nonpathogenic viruses as vaccine candidates.


Assuntos
Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A/imunologia , Vírus da Influenza A/patogenicidade , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Surtos de Doenças , Cães , Feminino , Humanos , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/isolamento & purificação , Vacinas contra Influenza/imunologia , Influenza Humana/epidemiologia , Influenza Humana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Vacinas de Produtos Inativados/imunologia
16.
Invest Ophthalmol Vis Sci ; 39(10): 1854-62, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9727408

RESUMO

PURPOSE: Neutrophils are the most prominent cell type to migrate initially into the herpes simplex virus type 1 (HSV-1)-infected murine cornea. The role the C-X-C chemokines macrophage inflammatory protein (MIP)-2 and KC play in promoting this response was investigated. METHODS: MIP-2 and KC were quantitated by enzyme-linked immunosorbent assay. Neutralization of endogenous MIP-2 and KC was achieved by subconjunctival inoculation of the appropriate antibody. Infected corneas were examined immunohistochemically for infiltrating leukocytes and assayed for myeloperoxidase activity using the dye o-dianisidine. Depletion of neutrophils and natural killer cells was accomplished by intraperitoneal administration of RB6-8C5 and asialo GM1 antibodies. RESULTS: Herpes simplex virus type 1, when introduced intracorneally, stimulated the production of MIP-2 and KC, with peak synthesis occurring 48 hours after infection. Dose-response studies showed that exogenous MIP-2 was three to four times more potent than KC in attracting neutrophils as assessed by myeloperoxidase assay and immunohistochemical staining. Subconjunctival administration of neutralizing antibody to MIP-2 resulted in a sharp decrease in neutrophil infiltration and significantly reduced corneal opacity scores. In contrast, in vivo treatment with neutralizing antibody to KC did not suppress ocular inflammation. Additional studies indicated that MIP-2 and KC could be made by corneal epithelial cells and that production was promoted by interleukin (IL)-1. In vivo depletion of neutrophils sharply reduced MIP-2 levels but did not affect KC levels. CONCLUSIONS: Collectively, the results suggest that MIP-2 is the major chemokine that attracts neutrophils into the HSV-1 infected cornea, where the cells directly or indirectly cause tissue injury. Resident corneal cells and inflammatory cells contribute to MIP-2 synthesis, whereas KC production seems to be confined largely to corneal cells.


Assuntos
Quimiotaxia de Leucócito , Córnea/metabolismo , Herpesvirus Humano 1 , Peptídeos e Proteínas de Sinalização Intercelular , Ceratite Herpética/metabolismo , Proteínas Inflamatórias de Macrófagos/fisiologia , Neutrófilos/fisiologia , Animais , Quimiocina CCL4 , Quimiocina CXCL1 , Quimiocinas CXC/fisiologia , Fatores Quimiotáticos/fisiologia , Córnea/patologia , Córnea/virologia , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Feminino , Substâncias de Crescimento/fisiologia , Interleucina-1/metabolismo , Interleucina-1/farmacologia , Ceratite Herpética/patologia , Células Matadoras Naturais/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Peroxidase/metabolismo
17.
J Leukoc Biol ; 63(4): 486-92, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9544579

RESUMO

Herpes simplex virus type 1 (HSV-1) infection of the murine cornea results in a tissue-destructive inflammatory response. In this study we show that virus infection induces the synthesis of macrophage inflammatory protein-2 (MIP-2), MIP-1alpha, and monocyte chemoattractant protein-1 (MCP-1). However, only the production of MIP-2 and MIP-1alpha coincided with the influx of leukocytes into the cornea. IL-10 treatment markedly suppressed chemokine message and protein synthesis in vivo. Local administration of IL-10 also dramatically reduced the number of T cells and neutrophils migrating into the cornea and suppressed the severity of corneal disease. The inflammatory response could also be suppressed by the passive transfer of neutralizing antibody to MIP-1alpha but not MCP-1. We conclude that local IL-10 administration can suppress chemokine synthesis, thereby ameliorating corneal disease. Furthermore, our results indicate that MIP-1alpha plays a major role in herpes stromal keratitis development, whereas MCP-1 does not.


Assuntos
Quimiocinas/metabolismo , Córnea/metabolismo , Herpesvirus Humano 1/imunologia , Interleucina-10/farmacologia , Ceratite Herpética/metabolismo , Animais , Anticorpos/farmacologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/imunologia , Quimiocina CCL2/fisiologia , Quimiocina CCL3 , Quimiocina CCL4 , Quimiocina CXCL2 , Quimiotaxia de Leucócito/efeitos dos fármacos , Córnea/efeitos dos fármacos , Feminino , Imunização Passiva , Imuno-Histoquímica , Proteínas Inflamatórias de Macrófagos/imunologia , Proteínas Inflamatórias de Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Monocinas/fisiologia , Reação em Cadeia da Polimerase , Fatores de Tempo
18.
J Virol ; 72(5): 3705-10, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9557652

RESUMO

Prior studies in our laboratory have suggested that the CC chemokine macrophage inflammatory protein-1alpha (MIP-1alpha) may be an important mediator in the blinding ocular inflammation which develops following herpes simplex virus type 1 (HSV-1) infection of the murine cornea. To directly test this hypothesis, MIP-1alpha-deficient (-/-) mice and their wild-type (+/+) counterparts were infected topically on the scarified cornea with 2.5 x 10(5) PFU of HSV-1 strain RE and subsequently graded for corneal opacity. Four weeks postinfection (p.i.), the mean corneal opacity score of -/- mice was 1.1 +/- 0.3 while that of the +/+ mice was 3.7 +/- 0.5. No detectable infiltrating CD4+ T cells were seen histologically at 14 or 21 days p.i. in -/- animals, whereas the mean CD4+ T-cell count per field (36 fields counted) in +/+ hosts was 26 +/- 2 (P < 0.001). In addition, neutrophil counts in the -/- mouse corneas were reduced by >80% in comparison to the wild-type controls. At 2 weeks p.i., no interleukin-2 or gamma interferon could be detected in six of seven -/- mice, whereas both T-cell cytokines were readily demonstrable in +/+ mouse corneas. Also, MIP-2 and monocyte chemoattractant protein-1 protein levels were significantly lower in MIP-1alpha -/- mouse corneas than in +/+ host corneas, suggesting that MIP-1alpha directly, or more likely indirectly, influences the expression of other chemokines. Interestingly, despite the paucity of infiltrating cells, HSV-1 clearance from the eyes of -/- mice was not significantly different from that observed in +/+ hosts. We conclude that MIP-1alpha is not needed to control virus growth in the cornea but is essential for the development of severe stromal keratitis.


Assuntos
Cegueira/fisiopatologia , Herpes Simples/fisiopatologia , Ceratite Herpética/fisiopatologia , Proteínas Inflamatórias de Macrófagos/fisiologia , Animais , Cegueira/imunologia , Cegueira/patologia , Cegueira/virologia , Quimiocina CCL2/metabolismo , Quimiocina CCL3 , Quimiocina CCL4 , Quimiocina CXCL2 , Chlorocebus aethiops , Córnea/metabolismo , Modelos Animais de Doenças , Feminino , Herpes Simples/imunologia , Herpes Simples/patologia , Herpes Simples/virologia , Herpesvirus Humano 1/imunologia , Humanos , Interferon gama/metabolismo , Interleucina-2/metabolismo , Ceratite Herpética/imunologia , Ceratite Herpética/patologia , Ceratite Herpética/virologia , Proteínas Inflamatórias de Macrófagos/genética , Masculino , Camundongos , Monocinas/metabolismo , Células Vero
19.
J Virol ; 70(2): 898-904, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8551629

RESUMO

Herpes simplex virus type 1 (HSV-1) infection of the murine cornea induces the rapid infiltration of neutrophils. We investigated whether these cells could influence virus replication. BALB/c mice treated with monoclonal antibody (MAb) RB6-8C5 experienced a profound depletion of neutrophils in the bloodstream, spleen, and cornea. In these animals, virus titers in the eye were significantly higher than those in the immunoglobulin G-treated controls at 3 days postinfection. By day 9, virus was no longer detectable in the controls, whereas titers of 10(3) to 10(6) PFU were still present in the neutrophil-depleted hosts. Furthermore, virus spread more readily to the skin and brains of MAb RB6-8C5-treated animals, rendering them significantly more susceptible to HSV-1-induced blepharitis and encephalitis. Only 25% of the treated animals survived, whereas all of the controls lived. Although MAb RB6-8C5 treatment did not alter the CD4+ T-cell, B-cell, natural killer cell, or macrophage populations, the CD8+ T-cell population was partially reduced. Therefore, the experiments were repeated in severe combined immunodeficiency mice, which lack CD8+ T cells. Again virus growth was found to be significantly elevated in the eyes, trigeminal ganglia, and brains of the MAb RB6-8C5-treated hosts. These results strongly indicate that in both immunocompetent and immunodeficient mice, neutrophils play a significant role in helping to control the replication and spread of HSV-1 after corneal infection.


Assuntos
Herpesvirus Humano 1/imunologia , Ceratite Herpética/imunologia , Neutrófilos/imunologia , Replicação Viral , Animais , Anticorpos Monoclonais/imunologia , Blefarite/imunologia , Blefarite/virologia , Linfócitos T CD8-Positivos/imunologia , Chlorocebus aethiops , Olho/virologia , Feminino , Herpesvirus Humano 1/crescimento & desenvolvimento , Humanos , Ceratite Herpética/virologia , Contagem de Leucócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Células Vero
20.
J Interferon Cytokine Res ; 16(1): 35-40, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8640449

RESUMO

Corneas excised from normal BALB/c mice and incubated in vitro were analyzed for the production of "early-warning" cytokines via reverse transcription-polymerase chain reaction and ELISA. It was found that the trauma of excision stimulated rapid IL-1 alpha synthesis, with peak protein accumulation occurring at 6 h, whereas IL-6 synthesis was maximal at 18 h. Neither IL-1 beta protein nor message was detected at any point, and TNF-alpha synthesis never increased above constituted levels. Antibody neutralization of endogenous IL-1 alpha blocked IL-6 synthesis. Addition of exogenous IL-1 alpha induced IL-1 alpha and IL-6 synthesis in vitro. Inoculation of IL-1 alpha into the cornea induced IL-6 synthesis in vivo. Addition of IL-1 alpha could stimulate IL-1R, IL-1 alpha, and IL-6 mRNA synthesis in the epithelial, stromal, and endothelial components of the cornea. However, protein production was readily detected only in the epithelial layer. We concluded that mechanical trauma to the mouse cornea triggers the enhanced synthesis of IL-1 alpha and IL-1R, which in turn results in the production of IL-6 and more IL-1 alpha. That corneal excision did not stimulate the synthesis of IL-1 beta or TNF-alpha indicates that there is a selective induction of early cytokine expression in this specialized tissue.


Assuntos
Córnea/metabolismo , Citocinas/biossíntese , RNA Mensageiro/biossíntese , Animais , Sequência de Bases , Lesões da Córnea , Citocinas/genética , Feminino , Técnicas In Vitro , Interleucina-1/biossíntese , Interleucina-6/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Fator de Necrose Tumoral alfa/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...