Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Proteome Res ; 14(2): 664-75, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25494920

RESUMO

Francisella tularensis causes disease (tularemia) in a large number of mammals, including man. We previously demonstrated enhanced efficacy of conventional antibiotic therapy for tularemia by postexposure passive transfer of immune sera developed against a F. tularensis LVS membrane protein fraction (MPF). However, the protein composition of this immunogenic fraction was not defined. Proteomic approaches were applied to define the protein composition and identify the immunogens of MPF. MPF consisted of at least 299 proteins and 2-D Western blot analyses using sera from MPF-immunized and F. tularensis LVS-vaccinated mice coupled to liquid chromatography-tandem mass spectrometry identified 24 immunoreactive protein spots containing 45 proteins. A reverse vaccinology approach that applied labeling of F. tularensis LVS surface proteins and bioinformatics was used to reduce the complexity of potential target immunogens. Bioinformatics analyses of the immunoreactive proteins reduced the number of immunogen targets to 32. Direct surface labeling of F. tularensis LVS resulted in the identification of 31 surface proteins. However, only 13 of these were reactive with MPF and/or F. tularensis LVS immune sera. Collectively, this use of orthogonal proteomic approaches reduced the complexity of potential immunogens in MPF by 96% and allowed for prioritization of target immunogens for antibody-based immunotherapies against tularemia.


Assuntos
Proteínas de Bactérias/metabolismo , Vacinas Bacterianas/uso terapêutico , Francisella tularensis/metabolismo , Proteínas de Membrana/metabolismo , Tularemia/prevenção & controle , Animais , Cromatografia Líquida , Mesotelina , Camundongos , Profilaxia Pós-Exposição , Espectrometria de Massas em Tandem
2.
Infect Immun ; 81(12): 4626-34, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24101688

RESUMO

Infections with the Gram-negative bacterium Burkholderia pseudomallei (melioidosis) are associated with high mortality, and there is currently no approved vaccine to prevent the development of melioidosis in humans. Infected patients also do not develop protective immunity to reinfection, and some individuals will develop chronic, subclinical infections with B. pseudomallei. At present, our understanding of what constitutes effective protective immunity against B. pseudomallei infection remains incomplete. Therefore, we conducted a study to elucidate immune correlates of vaccine-induced protective immunity against acute B. pseudomallei infection. BALB/c and C57BL/6 mice were immunized subcutaneously with a highly attenuated, Select Agent-excluded purM deletion mutant of B. pseudomallei (strain Bp82) and then subjected to intranasal challenge with virulent B. pseudomallei strain 1026b. Immunization with Bp82 generated significant protection from challenge with B. pseudomallei, and protection was associated with a significant reduction in bacterial burden in lungs, liver, and spleen of immunized mice. Humoral immunity was critically important for vaccine-induced protection, as mice lacking B cells were not protected by immunization and serum from Bp82-vaccinated mice could transfer partial protection to nonvaccinated animals. In contrast, vaccine-induced protective immunity was found to be independent of both CD4 and CD8 T cells. Tracking studies demonstrated uptake of the Bp82 vaccine strain predominately by neutrophils in vaccine-draining lymph nodes and by smaller numbers of dendritic cells (DC) and monocytes. We concluded that protection following cutaneous immunization with a live attenuated Burkholderia vaccine strain was dependent primarily on generation of effective humoral immune responses.


Assuntos
Anticorpos Antibacterianos/imunologia , Vacinas Bacterianas/imunologia , Burkholderia pseudomallei/imunologia , Melioidose/imunologia , Vacinas Atenuadas/imunologia , Animais , Vacinas Bacterianas/administração & dosagem , Burkholderia pseudomallei/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Feminino , Imunidade Humoral , Imunização , Melioidose/microbiologia , Melioidose/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Vacinação
3.
Infect Immun ; 81(9): 3099-105, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23774604

RESUMO

Francisella tularensis is a highly infectious intracellular bacterium that causes the zoonotic infection tularemia. While much literature exists on the host response to F. tularensis infection, the vast majority of work has been conducted using attenuated strains of Francisella that do not cause disease in humans. However, emerging data indicate that the protective immune response against attenuated F. tularensis versus F. tularensis type A differs. Several groups have recently reported that interleukin-17 (IL-17) confers protection against the live vaccine strain (LVS) of Francisella. While we too have found that IL-17Rα(-/-) mice are more susceptible to F. tularensis LVS infection, our studies, using a virulent type A strain of F. tularensis (SchuS4), indicate that IL-17Rα(-/-) mice display organ burdens and pulmonary gamma interferon (IFN-γ) responses similar to those of wild-type mice following infection. In addition, oral LVS vaccination conferred equivalent protection against pulmonary challenge with SchuS4 in both IL-17Rα(-/-) and wild-type mice. While IFN-γ was found to be critically important for survival in a convalescent model of SchuS4 infection, IL-17 neutralization from either wild-type or IFN-γ(-/-) mice had no effect on morbidity or mortality in this model. IL-17 protein levels were also higher in the lungs of mice infected with the LVS rather than F. tularensis type A, while IL-23p19 mRNA expression was found to be caspase-1 dependent in macrophages infected with LVS but not SchuS4. Collectively, these results demonstrate that IL-17 is dispensable for host immunity to type A F. tularensis infection, and that induced and protective immunity differs between attenuated and virulent strains of F. tularensis.


Assuntos
Vacinas Bacterianas/imunologia , Vacinas Bacterianas/farmacologia , Francisella tularensis/imunologia , Interleucina-17/genética , Interleucina-17/imunologia , Tularemia/genética , Tularemia/microbiologia , Animais , Caspase 1/genética , Caspase 1/imunologia , Feminino , Francisella tularensis/patogenicidade , Interferon gama/genética , Interferon gama/imunologia , Subunidade p19 da Interleucina-23/genética , Subunidade p19 da Interleucina-23/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores de Interleucina-17/imunologia , Tularemia/imunologia , Tularemia/prevenção & controle , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/farmacologia
4.
Vaccine ; 30(33): 4977-82, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22652404

RESUMO

Successful treatment of pneumonic infection with Francisella tularensis, the causative agent of tularemia, requires rapid initiation of antibiotic therapy, yet even then treatment failures may occur. Consequently, new treatments are needed to enhance the effectiveness of antimicrobial therapy for acute pneumonic tularemia. In a prior study, immunization with F. tularensis membrane protein fraction (MPF) antigens 3 days prior to challenge was reported to induce significant protection from inhalational challenge. We therefore hypothesized that MPF immunization might also be effective in enhancing infection control if combined with antibiotic therapy and administered after infection as post-exposure immunotherapy. To address this question, a 24h post-exposure treatment model of acute pulmonary Schu S4 strain of F. tularensis infection in BALB/c mice was used. Following exposure, mice were immunized with MPF and treated with low-dose gentamicin, alone or in combination and the effects on survival, bacterial burden and dissemination were assessed. We found that immunization with MPF significantly increased the effectiveness of subtherapeutic gentamicin for post-exposure treatment of pneumonic tularemia, with 100% of combination-treated mice surviving long-term. Bacterial burdens in the liver and spleen were significantly reduced in combination MPF-gentamicin treated mice at 7 days after challenge. Passively transferred antibodies against MPF antigens also increased the effectiveness of gentamicin therapy. Thus, we concluded that post-exposure immunization with MPF antigens was an effective means of enhancing conventional antimicrobial therapy for pneumonic tularemia.


Assuntos
Antibacterianos/administração & dosagem , Antibioticoprofilaxia/métodos , Vacinas Bacterianas/administração & dosagem , Gentamicinas/administração & dosagem , Profilaxia Pós-Exposição/métodos , Tularemia/prevenção & controle , Vacinação/métodos , Animais , Carga Bacteriana , Vacinas Bacterianas/imunologia , Modelos Animais de Doenças , Feminino , Francisella tularensis/imunologia , Fígado/microbiologia , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/imunologia , Mesotelina , Camundongos , Camundongos Endogâmicos BALB C , Baço/microbiologia , Análise de Sobrevida , Resultado do Tratamento
5.
Diagn Microbiol Infect Dis ; 64(2): 229-32, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19345041

RESUMO

Latex agglutination has been used to detect capsular polysaccharides from a variety of bacteria in body fluids. A latex agglutination assay was constructed for detection of the poly-gamma-D-glutamic acid (gammaDPGA) capsular polypeptide of Bacillus anthracis in serum from animal models of pulmonary anthrax. The assay was able to detect gammaDPGA in serum from infected animals at concentrations of 100 to 200 ng/mL.


Assuntos
Antraz/diagnóstico , Antígenos de Bactérias/análise , Bacillus anthracis/isolamento & purificação , Cápsulas Bacterianas/química , Ácido Poliglutâmico/análogos & derivados , Animais , Antígenos de Bactérias/imunologia , Bacillus anthracis/química , Bacillus anthracis/imunologia , Cápsulas Bacterianas/imunologia , Testes de Fixação do Látex/métodos , Camundongos , Camundongos Endogâmicos BALB C , Ácido Poliglutâmico/análise , Ácido Poliglutâmico/imunologia , Soro/química
6.
Infect Immun ; 77(1): 532-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19001075

RESUMO

Bacillus anthracis is surrounded by a capsular polypeptide composed of poly-gamma-D-glutamic acid (PGA). This antiphagocytic capsule is an essential virulence factor and is shed into body fluids during a murine model of pulmonary anthrax. Our previous studies of a murine model for antigen clearance showed that purified PGA accumulates in the liver and spleen, most notably in splenic macrophages and the Kupffer cells and sinusoidal endothelial cells of the liver. Although the tissue and cellular depots have been identified, there is little known about the uptake and intracellular fate of PGA. As a consequence, we examined the cellular uptake and organelle localization of PGA in the murine macrophage-like cell line J774.2. We found that PGA binds to and is internalized by J774.2 cells and accumulates in CD71 transferrin receptor-positive endosomes. The receptor-mediated endocytosis inhibitors amantadine and phenylarsine oxide inhibited the binding and uptake of PGA in these cells. Cytochalasin D and vinblastine, actin and microtubule inhibitors, respectively, failed to completely inhibit binding and uptake. Finally, we found that PGA is degraded in J774.2 cells starting 4 h after uptake, with continued degradation occurring for at least 24 h. This degradation of PGA may explain the rapid clearance of PGA that is observed in vivo compared to the slow clearance noted with capsular polysaccharides.


Assuntos
Bacillus anthracis/imunologia , Cápsulas Bacterianas/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Ácido Poliglutâmico/metabolismo , Animais , Antígenos CD/análise , Antígenos de Diferenciação de Linfócitos B/análise , Linhagem Celular , Endocitose , Endossomos/química , Camundongos
7.
Infect Immun ; 76(3): 899-906, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18195035

RESUMO

Bacillus anthracis is surrounded by an antiphagocytic capsule composed of poly-gamma-d-glutamic acid (gammaDPGA). Bacterial and fungal capsular polysaccharides are shed into body fluids in large amounts during infection. The goal of our study was to examine the in vivo fate and distribution of the gammaDPGA capsular polypeptide. Mice were injected via the intravenous route with various amounts of purified gammaDPGA. Blood, urine, and various organs were harvested at different times after treatment. Sites of gammaDPGA accumulation were determined by immunoassay using monoclonal antibodies specific for gammaDPGA. The results showed that the liver and spleen were the primary sites for the accumulation of gammaDPGA. As found in previous studies of capsular polysaccharides, the Kupffer cells of the liver and splenic macrophages were sites for the cellular accumulation of gammaDPGA. Unlike capsular polysaccharides, the hepatic sinusoidal endothelial cells were also sites for gammaDPGA accumulation. gammaDPGA was rapidly cleared from serum and was excreted into the urine. gammaDPGA in the urine showed a reduced molecular size relative to native gammaDPGA. The results indicate that in vivo clearance of the polypeptide capsular antigen of B. anthracis shares several features with the clearance of capsular polysaccharides. Key differences between the in vivo behaviors of gammaDPGA and capsular polysaccharides include the accumulation of gammaDPGA in hepatic sinusoidal endothelial cells and a gammaDPGA clearance rate that was more rapid than the clearance reported for capsular polysaccharides.


Assuntos
Antígenos de Bactérias/metabolismo , Bacillus anthracis/imunologia , Cápsulas Bacterianas/metabolismo , Ácido Poliglutâmico/metabolismo , Animais , Células Endoteliais/química , Ensaio de Imunoadsorção Enzimática , Feminino , Meia-Vida , Cinética , Células de Kupffer/química , Fígado/química , Macrófagos/química , Camundongos , Camundongos Endogâmicos BALB C , Soro/química , Baço/química , Urina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA