Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Comp Med ; 67(3): 222-231, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28662751

RESUMO

Animal models are vital in understanding the transmission and pathogenesis of infectious organisms and the host immune response to infection. In addition, animal models are essential in vaccine and therapeutic drug development and testing. Prior to selecting an animal model to use when studying an infectious agent, the scientific team must determine that sufficient in vitro and ex vivo data are available to justify performing research in an animal model, that ethical considerations are addressed, and that the data generated from animal work will add useful information to the body of scientific knowledge. Once it is established that an animal should be used, the questions become 'Which animal model is most suitable?' and 'Which experimental design issues should be considered?' The answers to these questions take into account numerous factors, including scientific, practical, welfare, and regulatory considerations, which are the focus of this article.


Assuntos
Experimentação Animal , Projetos de Pesquisa , Bem-Estar do Animal , Animais , Doenças Transmissíveis , Modelos Animais de Doenças , Transmissão de Doença Infecciosa
2.
Infect Immun ; 81(3): 1008-17, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23319564

RESUMO

Bacillus cereus strains harboring a pXO1-like virulence plasmid cause respiratory anthrax-like disease in humans, particularly in welders. We developed mouse models for intraperitoneal as well as aerosol challenge with spores of B. cereus G9241, harboring pBCXO1 and pBC218 virulence plasmids. Compared to wild-type B. cereus G9241, spores with a deletion of the pBCXO1-carried protective antigen gene (pagA1) were severely attenuated, whereas spores with a deletion of the pBC218-carried protective antigen homologue (pagA2) were not. Anthrax vaccine adsorbed (AVA) immunization raised antibodies that bound and neutralized the pagA1-encoded protective antigen (PA1) but not the PA2 orthologue encoded by pagA2. AVA immunization protected mice against a lethal challenge with spores from B. cereus G9241 or B. cereus Elc4, a strain that had been isolated from a fatal case of anthrax-like disease. As the pathogenesis of B. cereus anthrax-like disease in mice is dependent on pagA1 and PA-neutralizing antibodies provide protection, AVA immunization may also protect humans from respiratory anthrax-like death.


Assuntos
Bacillus cereus/imunologia , Vacinas Bacterianas/imunologia , Infecções por Bactérias Gram-Positivas/prevenção & controle , Infecções Respiratórias/prevenção & controle , Animais , Feminino , Infecções por Bactérias Gram-Positivas/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Infecções Respiratórias/microbiologia
3.
J Infect Dis ; 206(7): 1050-8, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22896664

RESUMO

Nonpigmented Yersinia pestis (pgm) strains are defective in scavenging host iron and have been used in live-attenuated vaccines to combat plague epidemics. Recently, a Y. pestis pgm strain was isolated from a researcher with hereditary hemochromatosis who died from laboratory-acquired plague. We used hemojuvelin-knockout (Hjv(-/-)) mice to examine whether iron-storage disease restores the virulence defects of nonpigmented Y. pestis. Unlike wild-type mice, Hjv(-/-) mice developed lethal plague when challenged with Y. pestis pgm strains. Immunization of Hjv(-/-) mice with a subunit vaccine that blocks Y. pestis type III secretion generated protection against plague. Thus, individuals with hereditary hemochromatosis may be protected with subunit vaccines but should not be exposed to live-attenuated plague vaccines.


Assuntos
Hemocromatose/complicações , Vacina contra a Peste/administração & dosagem , Peste/prevenção & controle , Yersinia pestis/patogenicidade , Animais , Feminino , Proteínas Ligadas por GPI , Hemocromatose/genética , Proteína da Hemocromatose , Fígado/microbiologia , Fígado/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Viabilidade Microbiana , Peste/genética , Peste/imunologia , Baço/microbiologia , Baço/patologia , Vacinas Atenuadas/administração & dosagem , Vacinas de Subunidades Antigênicas/administração & dosagem , Virulência , Yersinia pestis/imunologia
4.
Infect Immun ; 80(4): 1572-82, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22252870

RESUMO

Current efforts to develop plague vaccines focus on LcrV, a polypeptide that resides at the tip of type III secretion needles. LcrV-specific antibodies block Yersinia pestis type III injection of Yop effectors into host immune cells, thereby enabling phagocytes to kill the invading pathogen. Earlier work reported that antibodies against Y. pestis LcrV cannot block type III injection by Yersinia enterocolitica strains and suggested that lcrV polymorphisms may provide for escape from LcrV-mediated plague immunity. We show here that polyclonal or monoclonal antibodies raised against Y. pestis KIM D27 LcrV (LcrV(D27)) bind LcrV from Y. enterocolitica O:9 strain W22703 (LcrV(W22703)) or O:8 strain WA-314 (LcrV(WA-314)) but are otherwise unable to block type III injection by Y. enterocolitica strains. Replacing the lcrV gene on the pCD1 virulence plasmid of Y. pestis KIM D27 with either lcrV(W22703) or lcrV(WA-314) does not affect the ability of plague bacteria to secrete proteins via the type III pathway, to inject Yops into macrophages, or to cause lethal plague infections in mice. LcrV(D27)-specific antibodies blocked type III injection by Y. pestis expressing lcrV(W22703) or lcrV(WA-314) and protected mice against intravenous lethal plague challenge with these strains. Thus, although antibodies raised against LcrV(D27) are unable to block the type III injection of Y. enterocolitica strains, expression of lcrV(W22703) or lcrV(WA-314) in Y. pestis did not allow these strains to escape LcrV-mediated plague protective immunity in the intravenous challenge model.


Assuntos
Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Peste/imunologia , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/imunologia , Yersinia enterocolitica/genética , Yersinia enterocolitica/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/metabolismo , Antígenos de Bactérias/química , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Sistemas de Secreção Bacterianos , Linhagem Celular , Células HeLa , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Peste/microbiologia , Vacina contra a Peste/imunologia , Polimorfismo de Nucleotídeo Único , Proteínas Citotóxicas Formadoras de Poros/química , Alinhamento de Sequência , Yersinia enterocolitica/classificação , Yersinia pestis/imunologia , Yersinia pestis/patogenicidade
5.
Vaccine ; 29(38): 6572-83, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21763383

RESUMO

Yersinia pestis causes plague, a disease with high mortality in humans that can be transmitted by fleabite or aerosol. A US Food and Drug Administration (FDA)-licensed plague vaccine is currently not available. Vaccine developers have focused on two subunits of Y. pestis: LcrV, a protein at the tip of type III secretion needles, and F1, the fraction 1 pilus antigen. F1-V, a hybrid generated via translational fusion of both antigens, is being developed for licensure as a plague vaccine. The rV10 vaccine is a non-toxigenic variant of LcrV lacking residues 271-300. Here we developed Current Good Manufacturing Practice (cGMP) protocols for rV10. Comparison of clinical grade rV10 with F1-V did not reveal significant differences in plague protection in mice, guinea pigs or cynomolgus macaques. We also developed cGMP protocols for rV10-2, a variant of rV10 with an altered affinity tag. Immunization with rV10-2 adsorbed to aluminum hydroxide elicited antibodies against LcrV and conferred pneumonic plague protection in mice, rats, guinea pigs, cynomolgus macaques and African Green monkeys. The data support further development of rV10-2 for FDA Investigational New Drug (IND) authorization review and clinical testing.


Assuntos
Vacina contra a Peste/administração & dosagem , Vacina contra a Peste/imunologia , Peste/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Hidróxido de Alumínio/administração & dosagem , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Modelos Animais de Doenças , Cobaias , Macaca , Camundongos , Camundongos Endogâmicos BALB C , Doenças dos Primatas/prevenção & controle , Ratos , Doenças dos Roedores/prevenção & controle , Análise de Sobrevida , Vacinação/métodos , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Yersinia pestis/genética , Yersinia pestis/imunologia
6.
Crit Care Med ; 39(9): 2113-20, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21572326

RESUMO

OBJECTIVE: The pathogenesis and the outcome of Pseudomonas aeruginosa ventilator-acquired pneumonia depend on the virulence factors displayed by the bacteria as well as the host response. Thus, quorum sensing, lipopolysaccharide, and type 3 secretion system have each individually been shown to be important virulence systems in laboratory reference strains. However, the relative contribution of these three factors to the in vivo pathogenicity of clinically relevant strains has never been studied. We analyzed the virulence of 56 nonclonal Pseudomonas aeruginosa strains isolated from critically ill patients with ventilator-acquired pneumonia. To avoid the variation of human immune response, we used a murine model of pneumonia. The aim was to determine which virulence factor was the most important. SETTING: Research laboratory of a university. SUBJECTS: Male adult BALB/c mice. INTERVENTIONS: In vitro, the phenotype of each strain was established as to the expression of quorum sensing-regulated factors (elastase and pyocyanin), type 3 secretion system exotoxin secretion (Exotoxin U, S and/or T, or "nonsecreting"), and lipopolysaccharide O-antigen serotype. Strain pathogenicity was evaluated in vivo in a mouse model of acute pneumonia through lung injury assessment by measuring alveolar-capillary barrier permeability to proteins, lung wet/dry weight ratio, and bacterial dissemination. Associations were then sought between virulence system phenotypes and levels of lung injury. MEASUREMENTS AND MAIN RESULTS: In univariate analysis, elastase production, O11 serotype, and type 3 secretion system exotoxin secretion were associated with increased lung injury and exotoxin U was linked to an increase risk of bacteremia. In multivariate analysis, we observed that type 3 secretion system exotoxin secretion and to a lesser degree elastase production were associated with increased lung injury. CONCLUSION: In a murine model of pneumonia, our data suggest that type 3 secretion system and elastase are the most important virulence factors in clinically relevant P. aeruginosa strains.


Assuntos
Pneumonia Bacteriana/microbiologia , Pneumonia Associada à Ventilação Mecânica/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/patogenicidade , Fatores de Virulência/fisiologia , Animais , Bacteriemia/microbiologia , Proteínas de Bactérias/fisiologia , Modelos Animais de Doenças , Exotoxinas/fisiologia , Humanos , Masculino , Metaloendopeptidases/fisiologia , Camundongos , Camundongos Endogâmicos BALB C
7.
Am J Pathol ; 178(4): 1689-700, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21406168

RESUMO

Human pneumonic plague is a devastating and transmissible disease for which a Food and Drug Administration-approved vaccine is not available. Suitable animal models may be adopted as a surrogate for human plague to fulfill regulatory requirements for vaccine efficacy testing. To develop an alternative to pneumonic plague in nonhuman primates, we explored guinea pigs as a model system. On intranasal instillation of a fully virulent strain, Yersinia pestis CO92, guinea pigs developed lethal lung infections with hemorrhagic necrosis, massive bacterial replication in the respiratory system, and blood-borne dissemination to other organ systems. Expression of the Y. pestis F1 capsule was not required for the development of pulmonary infection; however, the capsule seemed to be important for the establishment of bubonic plague. The mean lethal dose (MLD) for pneumonic plague in guinea pigs was estimated to be 1000 colony-forming units. Immunization of guinea pigs with the recombinant forms of LcrV, a protein that resides at the tip of Yersinia type III secretion needles, or F1 capsule generated robust humoral immune responses. Whereas LcrV immunization resulted in partial protection against pneumonic plague challenge with 250 MLD Y. pestis CO92, immunization with recombinant F1 did not. rV10, a vaccine variant lacking LcrV residues 271-300, elicited protection against pneumonic plague, which seemed to be based on conformational antibodies directed against LcrV.


Assuntos
Peste/prevenção & controle , Vacinas de Subunidades Antigênicas/uso terapêutico , Animais , Modelos Animais de Doenças , Feminino , Cobaias , Humanos , Sistema Imunitário , Pulmão/microbiologia , Peste/fisiopatologia , Vacina contra a Peste/uso terapêutico , Conformação Proteica , Proteínas Recombinantes/química , Baço/microbiologia , Vacinas Sintéticas/química , Yersinia pestis/metabolismo
8.
Vaccine ; 28(7): 1870-6, 2010 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-20005318

RESUMO

LcrV, a protein that resides at the tip of the type III secretion needles of Yersinia pestis, is the single most important plague protective antigen. Earlier work reported monoclonal antibody MAb 7.3, which binds a conformational epitope of LcrV and protects experimental animals against lethal plague challenge. By screening monoclonal antibodies directed against LcrV for their ability to protect immunized mice against bubonic plague challenge, we examined here the possibility of additional protective epitopes. MAb BA5 protected animals against plague, neutralized the Y. pestis type III secretion pathway and promoted opsonophagocytic clearance of bacteria in blood. LcrV residues 196-225 were necessary and sufficient for MAb BA5 binding. Compared to full-length LcrV, a variant lacking its residues 196-225 retained the ability of eliciting plague protection. These results identify LcrV residues 196-225 as a linear epitope that is recognized by the murine immune system to confer plague protection.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Bactérias/genética , Epitopos/imunologia , Vacina contra a Peste/imunologia , Peste/prevenção & controle , Proteínas Citotóxicas Formadoras de Poros/genética , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Anticorpos Monoclonais/uso terapêutico , Afinidade de Anticorpos , Mapeamento de Epitopos , Feminino , Células HeLa , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose , Peste/imunologia , Peste/microbiologia
9.
Hum Vaccin ; 5(12): 817-23, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19786842

RESUMO

Yersinia pestis is the causative agent of bubonic and pneumonic plague, human diseases with high mortality. Due to the microbe's ability to spread rapidly, plague epidemics present a serious public health threat. A search for prophylactic measures was initially based on historical reports of bubonic plague survivors and their apparent immunity. Due to safety and efficacy concerns, killed whole-cell preparations or live-attenuated plague vaccines are no longer considered in the United States. Vaccine developers have focused on specific subunits of plague bacteria. LcrV, a protein at the tip of type III secretion needles, and F1, the capsular pilus antigen, are both recognized as plague protective antigens. Antibodies against LcrV and F1 interfere with Y. pestis type III injection of host cells. While LcrV is absolutely essential for Y. pestis virulence, expression of F1 is dispensable for plague pathogenesis in small animals, non-human primates and presumably also in humans. Several subunit vaccines, for example rF1+rV (rYP002), rF1V or rV10, are being developed to generate plague protection in humans. Efficacy testing and licensure for human use requires the establishment of correlates for plague immunity.


Assuntos
Vacina contra a Peste/imunologia , Peste/prevenção & controle , Yersinia pestis/imunologia , Yersinia pestis/patogenicidade , Animais , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Modelos Animais de Doenças , Humanos , Proteínas Citotóxicas Formadoras de Poros/imunologia , Primatas , Roedores , Estados Unidos , Vacinas de Subunidades Antigênicas/imunologia
10.
Infect Immun ; 77(5): 1807-16, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19237527

RESUMO

Yersinia pestis is perhaps the most feared infectious agent due to its ability to cause epidemic outbreaks of plague disease in animals and humans with high mortality. Plague infections elicit strong humoral immune responses against the capsular antigen (fraction 1 [F1]) of Y. pestis, and F1-specific antibodies provide protective immunity. Here we asked whether Y. pestis generates mutations that enable bacterial escape from protective immunity and isolated a variant with an IS1541 insertion in caf1A encoding the F1 outer membrane usher. The caf1A::IS1541 insertion prevented assembly of F1 pili and provided escape from plague immunity via F1-specific antibodies without a reduction in virulence in mouse models of bubonic or pneumonic plague. F1-specific antibodies interfere with Y. pestis type III transport of effector proteins into host cells, an inhibitory effect that was overcome by the caf1A::IS1541 insertion. These findings suggest a model in which IS1541 insertion into caf1A provides for reversible changes in envelope structure, enabling Y. pestis to escape from adaptive immune responses and plague immunity.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Elementos de DNA Transponíveis , Peste/imunologia , Peste/microbiologia , Yersinia pestis/genética , Yersinia pestis/imunologia , Animais , Contagem de Colônia Microbiana , DNA Bacteriano/química , DNA Bacteriano/genética , Feminino , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Peste/prevenção & controle , Recombinação Genética , Análise de Sequência de DNA , Baço/microbiologia , Baço/patologia , Análise de Sobrevida , Virulência
11.
Infect Immun ; 76(12): 5588-97, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18794281

RESUMO

Vaccine and therapeutic strategies that prevent infections with Yersinia pestis have been sought for over a century. Immunization with live attenuated (nonpigmented) strains and immunization with subunit vaccines containing recombinant low-calcium-response V antigen (rLcrV) and recombinant F1 (rF1) antigens are considered effective in animal models. Current antiplague subunit vaccines in development for utilization in humans contain both antigens, either as equal concentrations of the two components (rF1 plus rLcrV) or as a fusion protein (rF1-rLcrV). Here, we show that immunization with either purified rLcrV (a protein at the tip of type III needles) or a variant of this protein, recombinant V10 (rV10) (lacking amino acid residues 271 to 300), alone or in combination with rF1, prevented pneumonic lesions and disease pathogenesis. In addition, passive immunization studies showed that specific antibodies of macaques immunized with rLcrV, rV10, or rF1, either alone or in combination, conferred protection against bubonic plague challenge in mice. Finally, we found that when we compared the reactivities of anti-rLcrV and anti-rV10 immune sera from cynomolgus macaques, BALB/c mice, and brown Norway rats with LcrV-derived peptides, rV10, but not rLcrV immune sera, lacked antibodies recognizing linear LcrV oligopeptides.


Assuntos
Antígenos de Bactérias/imunologia , Vacina contra a Peste/imunologia , Peste/prevenção & controle , Vacinas Sintéticas/imunologia , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Proteínas de Bactérias/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Pulmão/imunologia , Pulmão/patologia , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Doenças dos Macacos/imunologia , Doenças dos Macacos/prevenção & controle , Peste/imunologia , Peste/patologia , Proteínas Citotóxicas Formadoras de Poros/imunologia , Ratos , Proteínas Recombinantes/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Yersinia pestis
12.
Biomed Environ Sci ; 21(2): 103-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18548848

RESUMO

OBJECTIVE: Pseudomonas aeruginosa is a ubiquitous and opportunistic pathogen that uses the type III secretion system (TTSS) to inject effector proteins directly into the cytosol of target cells to subvert the host cell's functions. Specialized bacterial chaperones are required for effective secretion of some effectors. To identify the chaperone of ExoS, the representative effector secreted by the TTSS of P. aeruginosa, we analyzed the role of a postulated chaperone termed Orf1. METHODS: By allelic exchange, we constructed the mutant with the deletion of gene Orf1. Analysis of secreted and cell-associated fractions was performed by SDS-PAGE and Western blotting. Using strain expressing in trans Orf1, tagged by V5 polypeptide and histidine, protein-protein interaction was determined by affinity resin pull-down assay in combination with MALDI-TOF. The role of Orf1 in the expression of exoS was evaluated by gene reporter analysis. RESULTS: Pull-down assay showed that Orf1 binds to ExoS and ExoT. Secretion profile analysis showed that Orf1 was necessary for the optimal secretion of ExoS and ExoT. However, Orf1 had no effect on the expression of exoS. CONCLUSION: Orf1 is important for the secretion of ExoS probably by maintaining ExoS in a secretion-competent conformation. We propose to name Orf1 as SpcS for "specific Pseudomonas chaperone for ExoS".


Assuntos
ADP Ribose Transferases/metabolismo , Toxinas Bacterianas/metabolismo , Chaperonas Moleculares/metabolismo , Pseudomonas aeruginosa/metabolismo , ADP Ribose Transferases/genética , Toxinas Bacterianas/genética , Sequência de Bases , Western Blotting , Primers do DNA , Eletroforese em Gel de Poliacrilamida , Cinética , Chaperonas Moleculares/genética , Ligação Proteica , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
13.
Infect Immun ; 76(5): 2025-36, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18347051

RESUMO

Yersinia pestis, the highly virulent agent of plague, is a biological weapon. Strategies that prevent plague have been sought for centuries, and immunization with live, attenuated (nonpigmented) strains or subunit vaccines with F1 (Caf1) antigen is considered effective. We show here that immunization with live, attenuated strains generates plague-protective immunity and humoral immune responses against F1 pilus antigen and LcrV. Y. pestis variants lacking caf1 (F1 pili) are not only fully virulent in animal models of bubonic and pneumonic plague but also break through immune responses generated with live, attenuated strains or F1 subunit vaccines. In contrast, immunization with purified LcrV, a protein at the tip of type III needles, generates protective immunity against the wild-type and the fully virulent caf1 mutant strain, in agreement with the notion that LcrV can elicit vaccine protection against both types of virulent plague strains.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Vacina contra a Peste/imunologia , Peste/prevenção & controle , Proteínas Citotóxicas Formadoras de Poros/imunologia , Yersinia pestis/genética , Yersinia pestis/imunologia , Animais , Anticorpos Antibacterianos/sangue , Proteínas de Bactérias/genética , Contagem de Colônia Microbiana , Feminino , Deleção de Genes , Imunoglobulina G/sangue , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Baço/microbiologia , Análise de Sobrevida , Vacinas Atenuadas/imunologia , Virulência
14.
Adv Exp Med Biol ; 603: 415-24, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17966437

RESUMO

Plague, an infectious disease that reached catastrophic proportions during three pandemics, continues to be a legitimate public health concern worldwide. Although antibiotic therapy for the causative agent Yersinia pestis is available, pharmaceutical supply limitations, multi-drug resistance from natural selection as well as malicious bioengineering are a reality. Consequently, plague vaccinology is a priority for biodefense research. Development of a multi-subunit vaccine with Fraction 1 and LcrV as protective antigens seems to be receiving the most attention. However, LcrV has been shown to cause immune suppression and Y. pestis mutants lacking F1 expression are thought to be fully virulent in nature and in animal experiments. The LcrV variant, rV10, retains the well documented protective antigenic properties of LcrV but with diminished inhibitory effects on the immune system. More research is required to examine the molecular mechanisms of vaccine protection afforded by surface protein antigens and to decipher the host mechanisms responsible for vaccine success.


Assuntos
Peste/imunologia , Peste/prevenção & controle , Animais , Proteínas da Membrana Bacteriana Externa/imunologia , Fímbrias Bacterianas/imunologia , Saúde Global , História do Século XX , História do Século XXI , Humanos , Peste/história , Vacina contra a Peste/história , Vacina contra a Peste/isolamento & purificação , Pesquisa/história , Vacinas de Subunidades Antigênicas/história , Vacinas de Subunidades Antigênicas/isolamento & purificação , Yersinia pestis/imunologia , Yersinia pestis/patogenicidade
15.
Mol Microbiol ; 65(2): 386-400, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17587230

RESUMO

Yersinia enterocolitica uses type III secretion to transport Yop proteins into the cytoplasm of host cells. Previous work generated hypotheses for both co- and post-translational transport mechanisms in the Yersinia type III pathway. Here, we used ubiquitin (Ub) and UBP1, the Ub-specific protease, to examine whether Yops can be secreted when synthesized prior to recognition by the type III machinery. Fusion of Ub to the N-terminus of Yops blocked substrate recognition and secretion of hybrids generated with YopE, YopQ or YopR. UBP1 removed Ub from the N-terminus of these hybrids and allowed YopE, YopQ or YopR cleavage products to enter the secretion pathway. Following the release of Ub, Yersinia type III machines also transported the YopE cleavage product into the cytosol of tissue culture cells. Minimal secretion signals were also examined with the Ub/UBP1 system and some, but not all, of these signals promoted type III secretion even after polypeptides had been freed from Ub. These results suggest that recognition and secretion of Yop substrates by the type III machinery can occur by a post-translational mechanism.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Sondas Moleculares/metabolismo , Sinais Direcionadores de Proteínas , Proteínas Recombinantes de Fusão/metabolismo , Ubiquitina/metabolismo , Yersinia enterocolitica/metabolismo , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Endopeptidases/metabolismo , Sondas Moleculares/química , Sondas Moleculares/genética , Peptídeos/metabolismo , Biossíntese de Proteínas , Transporte Proteico , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Ubiquitina/química , Ubiquitina/genética
16.
Mol Ther ; 14(5): 656-61, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17010670

RESUMO

Immunotherapy requiring an efficient T lymphocyte response is initiated by antigen delivery to antigen-presenting cells. Several studies have assessed the efficiency of various antigen loading procedures, including microbial vectors. Here a live strain of Pseudomonas aeruginosa was engineered to translocate a recombinant antigenic protein into mammalian cells via the type III secretion system, a bacterial device translocating effector proteins into host cells. Optimization of the vector included virulence attenuation and determination of the N-terminal sequence allowing translocation of fused antigens into cells. In vitro delivery of an ovalbumin fragment by the bacterial vector into dendritic cells induced the activation of ovalbumin-specific CD8(+) T lymphocytes. Mice injected with the ovalbumin-delivering vector developed ovalbumin-specific CD8(+) T lymphocytes and were resistant to a subsequent challenge with an ovalbumin-expressing melanoma. Moreover, in a curative assay, injection of the vaccine vector 5 and 12 days after tumor implantation led to a complete cure in five of six animals. These results highlight the utility of type III secretion system-based vectors for anti-tumor immunotherapy.


Assuntos
Antígenos/imunologia , Vetores Genéticos/genética , Imunoterapia , Neoplasias/genética , Neoplasias/imunologia , Pseudomonas aeruginosa/genética , Animais , Células Cultivadas , Células Dendríticas/imunologia , Engenharia Genética , Terapia Genética , Camundongos , Neoplasias/terapia
17.
Biotechniques ; 38(1): 63-7, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15679087

RESUMO

The complete genome of the bacterial pathogen Pseudomonas aeruginosa has now been sequenced, allowing gene deletion, one of the most frequently used methods in gene function study, to be fully exploited. In this study, we combine the sacB-based negative selection system with a cre-lox antibiotic marker recycling method. This methodology allows allelic exchange between a target gene and a gentamicin cassette flanked by the two lox sequences. A tetracycline plasmid expressing the cre recombinase is then introduced in the mutant strain to catalyze the excision of the lox-flanked resistance marker. We demonstrate here the efficiency of the combination of these two methods in P. aeruginosa by successively deleting ExoS and ExoT, which are two genetically independent toxins of the type-three secretion system (TTSS). This functional cre-lox recycling antibiotic marker system can create P. aeruginosa strains with multiple mutations without modifying the antibiotic resistance profile when compared to the parental strain.


Assuntos
Deleção de Genes , Marcação de Genes/métodos , Engenharia Genética/métodos , Marcadores Genéticos/genética , Integrases/genética , Pseudomonas aeruginosa/genética , Seleção Genética , Proteínas Virais/genética , Sequência de Bases , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Dados de Sequência Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...