Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Toxins (Basel) ; 16(6)2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38922165

RESUMO

Extensively drug-resistant Pseudomonas aeruginosa infections are emerging as a significant threat associated with adverse patient outcomes. Due to this organism's inherent properties of developing antibiotic resistance, we sought to investigate alternative strategies such as identifying "high value" antigens for immunotherapy-based purposes. Through extensive database mining, we discovered that numerous Gram-negative bacterial (GNB) genomes, many of which are known multidrug-resistant (MDR) pathogens, including P. aeruginosa, horizontally acquired the evolutionarily conserved gene encoding Zonula occludens toxin (Zot) with a substantial degree of homology. The toxin's genomic footprint among so many different GNB stresses its evolutionary importance. By employing in silico techniques such as proteomic-based phylogenetic tracing, in conjunction with comparative structural modeling, we discovered a highly conserved intermembrane associated stretch of 70 amino acids shared among all the GNB strains analyzed. The characterization of our newly identified antigen reveals it to be a "high value" vaccine candidate specific for P. aeruginosa. This newly identified antigen harbors multiple non-overlapping B- and T-cell epitopes exhibiting very high binding affinities and can adopt identical tertiary structures among the least genetically homologous P. aeruginosa strains. Taken together, using proteomic-driven reverse vaccinology techniques, we identified multiple "high value" vaccine candidates capable of eliciting a polarized immune response against all the P. aeruginosa genetic variants tested.


Assuntos
Filogenia , Infecções por Pseudomonas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/imunologia , Pseudomonas aeruginosa/genética , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Humanos , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/química , Vacinas contra Pseudomonas/imunologia , Vacinas contra Pseudomonas/genética , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito B/genética
2.
Biol Pharm Bull ; 43(10): 1469-1475, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32779581

RESUMO

Hospital-acquired infections with Pseudomonas aeruginosa have become a great challenge in caring for critically ill and immunocompromised patients. The cause of high mortality is the presence of multi-drug resistant (MDR) strains, which confers a pressing need for vaccines. Although vaccines against P. aeruginosa have been in development for more than several decades, there is no vaccine for patients at present. In this study, we purified genomic DNA of P. aeruginosa from sera of patients affected, constructed genome-wide library with random recombinants, and screened candidate protein antigens by evaluating their protective effects in vivo. After 13-round of screening, 115 reactive recombinants were obtained, among which 13 antigens showed strong immunoreactivity (more than 10% reaction to PcrV, a well-characterized V-antigen of P. aeruginosa). These 13 antigens were: PpiA, PtsP, OprP, CAZ10_34235, HmuU_2, PcaK, CarAd, RecG, YjiR_5, LigD, KinB, RtcA, and PscF. In vivo studies showed that vaccination with PscF protected against lethal P. aeruginosa challenge, and decreased lung inflammation and injury. A genomic library of P. aeruginosa could be constructed in this way for the first time, which could not only screen candidate antigens but also in a high-throughput way. PscF was considered as an ideal promising vaccine candidate for combating P. aeruginosa infection and was supported for further evaluation of its safety and efficacy.


Assuntos
Antígenos de Bactérias/genética , Infecção Hospitalar/prevenção & controle , Infecções por Pseudomonas/prevenção & controle , Vacinas contra Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/isolamento & purificação , Infecção Hospitalar/sangue , Infecção Hospitalar/imunologia , Infecção Hospitalar/microbiologia , DNA Bacteriano/genética , Modelos Animais de Doenças , Feminino , Biblioteca Genômica , Ensaios de Triagem em Larga Escala/métodos , Humanos , Imunogenicidade da Vacina , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Camundongos , Infecções por Pseudomonas/sangue , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Vacinas contra Pseudomonas/administração & dosagem , Vacinas contra Pseudomonas/genética , Pseudomonas aeruginosa/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
3.
Front Immunol ; 11: 601601, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33552056

RESUMO

Pulmonary infection caused by Pseudomonas aeruginosa (PA) has created an urgent need for an efficient vaccine, but the protection induced by current candidates is limited, partially because of the high variability of the PA genome. Antigens targeting pulmonary Th17 responses are able to provide antibody-independent and broad-spectrum protection; however, little information about Th17-stimulating antigens in PA is available. Herein, we identified two novel PA antigens that effectively induce Th17-dependent protection, namely, PcrV (PA1706) and AmpC (PA4110). Compared to intramuscular immunization, intranasal immunization enhanced the protection of rePcrV due to activation of a Th17 response. The Th17-stimulating epitopes of PcrV and AmpC were identified, and the recombinant protein PVAC was designed and generated by combining these Th17-stimulating epitopes. PVAC was successfully produced in soluble form and elicited broad protective immunity against PA. Our results provide an alternative strategy for the development of Th17-based vaccines against PA and other pathogens.


Assuntos
Antígenos de Bactérias , Proteínas de Bactérias , Toxinas Bacterianas , Epitopos de Linfócito T , Proteínas Citotóxicas Formadoras de Poros , Vacinas contra Pseudomonas , Pseudomonas aeruginosa , Células Th17/imunologia , beta-Lactamases , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Feminino , Camundongos , Camundongos Knockout , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/imunologia , Vacinas contra Pseudomonas/genética , Vacinas contra Pseudomonas/imunologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/imunologia , beta-Lactamases/genética , beta-Lactamases/imunologia
4.
Front Immunol ; 9: 1737, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30093906

RESUMO

Pseudomonas aeruginosa is a ubiquitous opportunistic pathogen, which causes infectious disease in patients with cystic fibrosis and compromised immunity. P. aeruginosa is difficult to eradicate because of its intrinsic resistance to most traditional antibiotics as well as acquired resistance mechanisms after decades of antibiotic usage. A full understanding of the P. aeruginosa pathogenesis mechanisms is necessary for the development of novel prevention and treatment strategies. To identify novel vaccine candidates, here we comprehensively examined the expression levels of all the known outer membrane proteins in two P. aeruginosa strains in a murine acute pneumonia model. OprH was one of the most highly expressed proteins during infection. In addition, OprH is known to be highly immunogenic and accessible by host proteins. Thus, it was chosen as a vaccine candidate. To further identify vaccine candidates, 34 genes highly expressed during infection were evaluated for their contributions in virulence by testing individual transposon insertion mutants. Among them, fpvA, hasR, and foxA were found essential for bacterial virulence and therefore included in vaccine construction. Immunization with a mixture of FpvA, HasR, and FoxA rendered no protection, however, while immunization by OprH refolded in liposomes elicited specific opsonic antibodies and conferred protection against two lipopolysaccharide-heterologous P. aeruginosa strains (PA14 and PA103). Overall, by studying the expression profile of the P. aeruginosa outer membrane proteins during infection, we identified OprH as a potential vaccine candidate for the prevention of lung infection by P. aeruginosa.


Assuntos
Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Lipopolissacarídeos/imunologia , Infecções por Pseudomonas/prevenção & controle , Vacinas contra Pseudomonas/imunologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Modelos Animais de Doenças , Escherichia coli/genética , Escherichia coli/imunologia , Feminino , Expressão Gênica , Humanos , Imunização , Camundongos , Fagocitose/imunologia , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/prevenção & controle , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Vacinas contra Pseudomonas/genética , Virulência
5.
J Microbiol Immunol Infect ; 51(3): 312-320, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28291719

RESUMO

BACKGROUND: Pseudomonas aeruginosa is a Gram-negative opportunistic bacterium, which considered as a common cause of nosocomial infection and life-threatening complications in immunocompromized and cystic fibrosis patients. Here, we evaluate the protective effect of recombinant vaccines composed of outer membrane proteins OprF and OprI alone or in combination with flagellin B against mucoid and nonmucoid pseudomonas infection. METHODS: BALB/C mice were immunized subcutaneous using OprF and OprI with or without flagellin B and antibody titers were determined. Serum bactericidal and opsonophagocytosis activities of immunized and control sera were estimated against mucoid and nonmucoid pseudomonas strains. Lung tissue sections from immunized and nonimmunized mice were analyzed and the levels of peripheral neutrophils infiltration into the lung and tissue inflammation were scored. RESULTS: Subcutaneous immunization using OprF and OprI with or without flagellin B elicited higher antibody titers against OprF, OprI, and flagellin B. The produced antibodies successfully opsonized both mucoid and nonmucoid strains with subsequent activation of the terminal pathway of complement that enhances killing of nonmucoid strains via complement-mediated lysis. Furthermore, opsonized mucoid and nonmucoid strains showed enhanced opsonophagocytosis via human peripheral neutrophils, a mechanism that kills P. aeruginosa when complement mediated lysis is not effective especially with mucoid strains. Immunized mice also showed a significant prolonged survival time, lower bacteremia, and reduced lung damage when compared with control nonimmunized mice. CONCLUSION: Our data showed that mice immunized with OprF/OprI or OprF/OprI and flagellin B are significantly protected from infection caused by mucoid and nonmucoid strains of P. aeruginosa.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Bactérias/imunologia , Flagelina/imunologia , Imunização , Lipoproteínas/imunologia , Infecções por Pseudomonas/prevenção & controle , Vacinas contra Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Infecções Respiratórias/prevenção & controle , Animais , Anticorpos Antibacterianos/sangue , Proteínas de Bactérias/genética , Clonagem Molecular , Modelos Animais de Doenças , Feminino , Flagelina/genética , Regulação Bacteriana da Expressão Gênica , Lipoproteínas/genética , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Vacinas contra Pseudomonas/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Infecções Respiratórias/microbiologia , Vacinação , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico
6.
Front Immunol ; 9: 3021, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30687303

RESUMO

High incidence, severity and increasing antibiotic resistance characterize Pseudomonas aeruginosa infections, highlighting the need for new therapeutic options. Vaccination strategies to prevent or limit P. aeruginosa infections represent a rational approach to positively impact the clinical outcome of risk patients; nevertheless this bacterium remains a challenging vaccine target. To identify novel vaccine candidates, we started from the genome sequence analysis of the P. aeruginosa reference strain PAO1 exploring the reverse vaccinology approach integrated with additional bioinformatic tools. The bioinformatic approaches resulted in the selection of 52 potential antigens. These vaccine candidates were conserved in P. aeruginosa genomes from different origin and among strains isolated longitudinally from cystic fibrosis patients. To assess the immune-protection of single or antigens combination against P. aeruginosa infection, a vaccination protocol was established in murine model of acute respiratory infection. Combinations of selected candidates, rather than single antigens, effectively controlled P. aeruginosa infection in the in vivo model of murine pneumonia. Five combinations were capable of significantly increase survival rate among challenged mice and all included PA5340, a hypothetical protein exclusively present in P. aeruginosa. PA5340 combined with PA3526-MotY gave the maximum protection. Both proteins were surface exposed by immunofluorescence and triggered a specific immune response. Combination of these two protein antigens could represent a potential vaccine to prevent P. aeruginosa infection.


Assuntos
Genoma Bacteriano , Genômica , Infecções por Pseudomonas/imunologia , Vacinas contra Pseudomonas/genética , Vacinas contra Pseudomonas/imunologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/imunologia , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Fibrose Cística/complicações , Modelos Animais de Doenças , Estudo de Associação Genômica Ampla , Genômica/métodos , Humanos , Camundongos , Fases de Leitura Aberta , Infecções por Pseudomonas/etiologia , Infecções por Pseudomonas/mortalidade , Infecções por Pseudomonas/prevenção & controle , Vacinas contra Pseudomonas/administração & dosagem , Pseudomonas aeruginosa/isolamento & purificação
7.
Microbiol Immunol ; 59(9): 533-44, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26249788

RESUMO

Pseudomonas aeruginosa poses a major threat to human health and to the mink industry. Thus, development of vaccines that elicit robust humoral and cellular immunity against P. aeruginosa is greatly needed. In this study, a recombinant attenuated Salmonella vaccine (RASV) that expresses the outer membrane proteins fusion OprF190-342 -OprI21-83 (F1I2) from P. aeruginosa was constructed and the potency of this vaccine candidate assessed by measuring F1I2-specific humoral immune responses upon vaccination through s.c. or oral routes. S.C. administration achieved higher serum IgG titers and IgA titers in the intestine and induced stronger F1I2-specific IgG and IgA titers in lung homogenate than did oral administration, which resulted in low IgG titers and no local IgA production. High titers of IFN-γ, IL-4, and T-lymphocyte subsets induced a mixed Th1/Th2 response in mice immunized s.c., indicating elicitation of cellular immunity. Importantly, when immunized mice were challenged with P. aeruginosa by the intranasal route 30 days after the initial immunization, s.c. vaccination achieved 77.78% protection, in contrast to 41.18% via oral administration and 66.67% via Escherichia coli-expressed F1I2 (His-F1I2) vaccination. These results indicate that s.c. vaccination provides a better protective response against P. aeruginosa infection than do oral administration and the His-F1I2 vaccine.


Assuntos
Proteínas de Bactérias/imunologia , Portadores de Fármacos , Lipoproteínas/imunologia , Infecções por Pseudomonas/prevenção & controle , Vacinas contra Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Salmonella typhimurium/genética , Administração Oral , Animais , Anticorpos Antibacterianos/análise , Anticorpos Antibacterianos/sangue , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Escherichia coli , Feminino , Imunoglobulina A/análise , Imunoglobulina G/sangue , Injeções Subcutâneas , Interferon gama/metabolismo , Interleucina-4/metabolismo , Mucosa Intestinal/imunologia , Lipoproteínas/genética , Pulmão/imunologia , Camundongos Endogâmicos BALB C , Vison , Vacinas contra Pseudomonas/administração & dosagem , Vacinas contra Pseudomonas/genética , Pseudomonas aeruginosa/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Vacinas contra Salmonella/administração & dosagem , Vacinas contra Salmonella/genética , Salmonella typhimurium/crescimento & desenvolvimento , Soro/imunologia , Análise de Sobrevida , Subpopulações de Linfócitos T/imunologia , Resultado do Tratamento , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia
8.
Hum Vaccin Immunother ; 10(1): 170-83, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24064511

RESUMO

INTRODUCTION: IC43 is a recombinant outer membrane protein-based vaccine against Pseudomonas aeruginosa (P. aeruginosa) consisting of OprF- and OprI- epitopes (Opr, outer membrane protein; OprF/I, OprF/OprI hybrid vaccine) with an N-terminal His 6 tag (Met-Ala-(His)6-OprF190-342-OprI21-83). OBJECTIVES: The study aimed to confirm the optimal dose of IC43 in adults with regard to immunogenicity, safety, and tolerability after vaccination with three different dosages of IC43, compared with placebo, and to investigate a potential immune-enhancing effect of the adjuvant, aluminum hydroxide. Subjects were randomly allocated in a 1:1:1:1:1 ratio to one of five treatment groups: 50, 100, or 200 µg IC43 with adjuvant, 100 µg IC43 without adjuvant, or placebo (0.9% sodium chloride) and two intramuscular injections were given in the deltoid region 7 d apart. RESULTS: The primary immunogenicity analysis of OprF/I-specific IgG antibody titers on day 14 demonstrated statistically significant differences among treatment groups (P<0.0001), with a significantly higher immune response detected in each IC43 treatment group compared with placebo. From day 0 to day 14, a ≥4-fold increase in OprF/I-specific immunoglobulin G (IgG) antibody titers were observed in>90% of subjects in all IC43 treatment groups in the per-protocol (PP) and intention-to-treat (ITT) populations; a ≥50-fold titer increase was observed in 42.6% subjects including all IC43 treatment groups. On day 90, OprF/I-specific IgGs started to decline in all IC43 treatment groups but remained significantly higher until 6 mo compared with placebo. Assessment of functional antibody induction by opsonophagocytic assay (OPA) followed a similar pattern compared with OprF/I-specific IgG kinetics. At day 14, a ≥2-fold increase in OPA titer was observed in 54.5% subjects within all IC43 treatment groups. An increase in antibody avidity index was observed after the second vaccination. At day 14, >96% of subjects in each IC43 treatment group had detectable OprF/I-specific IgG antibodies. Anti-histidine IgG antibody titers peaked on day 14 and were reduced on day 90 in all IC43 treatment groups. OprF/I-specific IgG secreted by antibody-secreting cell (ASC) was detected in all IC43 groups by B-cell ELIspot after the second vaccination and up to 6 mo. All vaccinations were safe and well tolerated up to the maximum cumulative dosage of 400 µg IC43. CONCLUSION: IC43 doses equal to or greater than 50 µg were sufficient to induce a plateau of IgG antibody responses in healthy volunteers. Higher doses, whether adjuvanted or non-adjuvanted, were not more effective. METHODS: In this phase I, randomized, placebo-controlled, observer-blinded, multicenter clinical trial, 163 healthy volunteers (18-65 y) were randomly assigned to five treatment groups (1:1:1:1:1). Three groups received IC43 with adjuvant: 50 µg (n=32), 100 µg (n=33), or 200 µg (n=33). One group received IC43 100 µg without adjuvant (n=32), and one group received placebo (0.9% sodium chloride) (n=33). Each subject received two intramuscular vaccinations, separated by a 7-d interval (days 0 and 7) (Fig. 1). Humoral immune response was assessed by measurement of outer membrane protein F/I (OprF/I)-specific antibodies determined by enzyme-linked immunosorbent assay (ELISA), anti-histidine antibodies determined by ELISA, and functional antibody activity determined by opsonophagocytic assay (OPA), up to 6 mo post-vaccination. Antibody avidity was measured on days 7 and 14 from samples that had detectable vaccine antibody-specific immunoglobulin G (IgG) antibody titers. At the Austrian site only, the B-cell ELIspot assay was used to determine specific ASC responses. Safety was assessed using adverse event monitoring and clinical laboratory tests. Local and systemic tolerability was recorded in a subject diary for 7 d after each vaccination and by investigators up to 6 mo post-vaccination.


Assuntos
Proteínas de Bactérias/imunologia , Lipoproteínas/imunologia , Vacinas contra Pseudomonas/efeitos adversos , Vacinas contra Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Adolescente , Adulto , Anticorpos Antibacterianos/sangue , Proteínas de Bactérias/genética , Feminino , Voluntários Saudáveis , Humanos , Imunoglobulina G/sangue , Injeções Intramusculares , Lipoproteínas/genética , Masculino , Pessoa de Meia-Idade , Placebos/administração & dosagem , Vacinas contra Pseudomonas/administração & dosagem , Vacinas contra Pseudomonas/genética , Pseudomonas aeruginosa/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Vacinação/efeitos adversos , Vacinação/métodos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Adulto Jovem
9.
Artigo em Russo | MEDLINE | ID: mdl-23297634

RESUMO

AIM: Study of biological properties of strains-producers of a number of Pseudomonas aeruginosa recombinant proteins compared with the initial strains. MATERIALS AND METHODS: 5 Escherichia coli strains (initial strains E. coli M15 and E. coli BL21 (DE3) and strains-producers of OprL, OprF, aTox3 recombinant proteins (E. coli M15/oprF, E. coli M 15/oprL, E. coli BL21-aTox3) were studied. Toxicity, toxigenicity and virulence determination were carried out in experiments in non-linear mice. Enzymatic properties of the initial strains and strains-producers were compared in a number of biochemical tests. RESULTS: The studied strains-producers of the most immunogenic P. aeruginosa recombinant proteins were confirmed to be biosafe, belong to Enterobacteriaceae family and Escherichia genus. As a result of genetic engineering manipulations carried out with E. coli M15 and E. coli BL21(DE3) alterations ofbiochemical and growth properties, virulence, toxicity and toxigenicity in the constructed strains-producers of recombinant proteins - E. coli M 15/oprF, E. coli M15/oprL, E. coli BL21-aTox3 - were not detected. CONCLUSION: The results obtained allow to consider the possibility of use of E. coli M 15/oprF, E. coli M15/oprL and E. coli BL21-aTox3 strains-producers for production and isolation of candidate proteins for inclusion into vaccine against pseudomonas infection.


Assuntos
Proteínas de Bactérias/biossíntese , Escherichia coli/genética , Escherichia coli/patogenicidade , Infecções por Pseudomonas/prevenção & controle , Vacinas contra Pseudomonas/biossíntese , Pseudomonas aeruginosa/imunologia , Proteínas Recombinantes/biossíntese , Animais , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/genética , Escherichia coli/metabolismo , Fermentação , Engenharia Genética , Camundongos , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Vacinas contra Pseudomonas/administração & dosagem , Vacinas contra Pseudomonas/genética , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/genética , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Virulência
10.
Vaccine ; 27(30): 4047-55, 2009 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-19501788

RESUMO

Ferric uptake regulator (Fur) is a global transcription regulator that is ubiquitous to Gram-negative bacteria and regulates diverse biological processes, including iron uptake, cellular metabolism, stress response, and production of virulence determinants. As a result, for many pathogenic bacteria, Fur plays a crucial role in the course of infection and disease development. In this study, the fur gene was cloned from a pathogenic Pseudomonas fluorescens strain, TSS, isolated from diseased Japanese flounder cultured in a local farm. TSS Fur can partially complement the defective phenotype of an Escherichia coli fur mutant. A TSS fur null mutant, TFM, was constructed. Compared to TSS, TFM exhibits reduced growth ability, aberrant production of outer membrane proteins, decreased resistance against host serum bactericidal activity, impaired ability to disseminate in host blood and tissues, and drastic attenuation in overall bacterial virulence in a Japanese flounder infection model. When used as a live vaccine administered via the injection, immersion, and oral routes, TFM affords high levels of protection upon Japanese flounder against not only P. fluorescens infection but also Aeromonas hydrophila infection. Furthermore, a plasmid, pJAQ, was constructed, which expresses the coding element of the Vibrio harveyi antigen AgaV-DegQ. TFM harboring pJAQ can secret AgaV-DegQ into the extracellular milieu. Vaccination of Japanese flounder with live TFM/pJAQ elicited strong immunoprotection against both V. harveyi and A. hydrophila infections.


Assuntos
Infecções por Bactérias Gram-Negativas/prevenção & controle , Vacinas contra Pseudomonas/imunologia , Vacinas contra Pseudomonas/microbiologia , Pseudomonas fluorescens/imunologia , Pseudomonas fluorescens/patogenicidade , Aeromonas hydrophila/imunologia , Animais , Proteínas de Bactérias/genética , Clonagem Molecular , DNA Bacteriano/química , DNA Bacteriano/genética , Escherichia coli/genética , Linguado/microbiologia , Deleção de Genes , Teste de Complementação Genética , Dados de Sequência Molecular , Plasmídeos , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Infecções por Pseudomonas/prevenção & controle , Vacinas contra Pseudomonas/genética , Pseudomonas fluorescens/genética , Pseudomonas fluorescens/isolamento & purificação , Proteínas Repressoras/genética , Análise de Sequência de DNA , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vibrioses/prevenção & controle , Virulência , Fatores de Virulência/genética
11.
Infect Immun ; 77(6): 2356-66, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19349426

RESUMO

Although chronic Pseudomonas aeruginosa infection is the major cause of morbidity and mortality in cystic fibrosis (CF) patients, there is no approved vaccine for human use against P. aeruginosa. The goal of this study was to establish whether a multivalent vaccine containing P. aeruginosa type A and B flagellins as well as the outer membrane proteins OprF and OprI would promote enhanced clearance of P. aeruginosa. Intramuscular immunization with flagellins and OprI (separate) or OprI-flagellin fusion proteins generated significant antiflagellin immunoglobulin G (IgG) responses. However, only the fusions of OprI with type A and type B flagellins generated OprI-specific IgG. Immunization with a combination of OprF epitope 8 (OprF(311-341)), OprI, and flagellins elicited high-affinity IgG antibodies specific to flagellins, OprI, and OprF that individually promoted extensive deposition of C3 on P. aeruginosa. Although these antibodies exhibited potent antibody-dependent complement-mediated killing of nonmucoid bacteria, they were significantly less effective with mucoid isolates. Mice immunized with the OprF(311-341)-OprI-flagellin fusion had a significantly lower bacterial burden three days postchallenge and cleared the infection significantly faster than control mice. In addition, mice immunized with the OprF(311-341)-OprI-flagellin fusion had significantly less inflammation and lung damage throughout the infection than OprF-OprI-immunized mice. Based on our results, OprF(311-341)-OprI-flagellin fusion proteins have substantial potential as components of a vaccine against nonmucoid P. aeruginosa, which appears to be the phenotype of the bacterium that initially colonizes CF patients.


Assuntos
Proteínas de Bactérias/imunologia , Epitopos/imunologia , Flagelina/imunologia , Lipoproteínas/imunologia , Vacinas contra Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Animais , Anticorpos Antibacterianos/sangue , Proteínas de Bactérias/genética , Contagem de Colônia Microbiana , Complemento C3/imunologia , Complemento C3/metabolismo , Epitopos/genética , Flagelina/genética , Humanos , Imunoglobulina G/sangue , Lipoproteínas/genética , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Viabilidade Microbiana , Infecções por Pseudomonas/imunologia , Vacinas contra Pseudomonas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 25(3): 193-5, 2009 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-19257978

RESUMO

AIM: To construct the MyD88-Pseudomonas aeruginosa epitope vaccine and study its expression in eukaryotic cells. METHODS: To design and synthesize an epigene containing three B cell epitopes of OprF and one foreign "promiscuous" T cell epitope by overlapping extension PCR. tPA signal encoding sequence was amplified by PCR and then it was inserted into the 5' terminus of the epigene to construct tPA-OprF. tPA-OprF and MyD88 were cloned into the expression vector pIRES and the recombinant plasmid pIRES-tPAOprF-MyD88 was constructed. The recombinant plasmid was transfected into COS-7 cells by electroporation. The expression protein of tPA-OprF and MyD88 was detected by Western blot. RESULTS: The recombinant plasmid pIRES-tPA-OprF-MyD88 was successfully constructed. Western blot analysis indicated the tPA-OprF fusion protein was expressed in supertanant of COS-7 cells and MyD88 protein in COS-7 cells. CONCLUSION: The recombinant plasmid pIRES-tPA-OprF-MyD88 has been successfully constructed and tPA-OprF and MyD88 protein can be highly expressed in transfected cells. It may be used as a potential candidate of preventive vaccine of pseudomonas aeruginosa.


Assuntos
Proteínas de Bactérias/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Vacinas contra Pseudomonas/metabolismo , Ativador de Plasminogênio Tecidual/metabolismo , Animais , Proteínas de Bactérias/genética , Western Blotting , Células COS , Chlorocebus aethiops , Clonagem Molecular , Humanos , Fator 88 de Diferenciação Mieloide/genética , Reação em Cadeia da Polimerase , Vacinas contra Pseudomonas/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Ativador de Plasminogênio Tecidual/genética , Transfecção , Vacinas de DNA/genética , Vacinas de DNA/metabolismo
13.
J Immunol ; 181(7): 4965-75, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18802100

RESUMO

In a murine model of acute fatal pneumonia, we previously showed that nasal immunization with a live-attenuated aroA deletant of Pseudomonas aeruginosa strain PAO1 elicited LPS serogroup-specific protection, indicating that opsonic Ab to the LPS O Ag was the most important immune effector. Because P. aeruginosa strain PA14 possesses additional virulence factors, we hypothesized that a live-attenuated vaccine based on PA14 might elicit a broader array of immune effectors. Thus, an aroA deletant of PA14, denoted PA14DeltaaroA, was constructed. PA14DeltaaroA-immunized mice were protected against lethal pneumonia caused not only by the parental strain but also by cytotoxic variants of the O Ag-heterologous P. aeruginosa strains PAO1 and PAO6a,d. Remarkably, serum from PA14DeltaaroA-immunized mice had very low levels of opsonic activity against strain PAO1 and could not passively transfer protection, suggesting that an antibody-independent mechanism was needed for the observed cross-serogroup protection. Compared with control mice, PA14DeltaaroA-immunized mice had more rapid recruitment of neutrophils to the airways early after challenge. T cells isolated from P. aeruginosa DeltaaroA-immunized mice proliferated and produced IL-17 in high quantities after coculture with gentamicin-killed P. aeruginosa. Six hours following challenge, PA14DeltaaroA-immunized mice had significantly higher levels of IL-17 in bronchoalveolar lavage fluid compared with unimmunized, Escherichia coli-immunized, or PAO1DeltaaroA-immunized mice. Antibody-mediated depletion of IL-17 before challenge or absence of the IL-17 receptor abrogated the PA14DeltaaroA vaccine's protection against lethal pneumonia. These data show that IL-17 plays a critical role in antibody-independent vaccine-induced protection against LPS-heterologous strains of P. aeruginosa in the lung.


Assuntos
Interleucina-17/fisiologia , Lipopolissacarídeos/imunologia , Pneumonia Bacteriana/prevenção & controle , Infecções por Pseudomonas/prevenção & controle , Vacinas contra Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Animais , Anticorpos Antibacterianos/biossíntese , Células Cultivadas , Feminino , Lipopolissacarídeos/classificação , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Antígenos O/classificação , Antígenos O/genética , Antígenos O/imunologia , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/mortalidade , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/mortalidade , Vacinas contra Pseudomonas/administração & dosagem , Vacinas contra Pseudomonas/genética , Sorotipagem , Baço/citologia , Baço/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Auxiliares-Indutores/microbiologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...