Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Hum Vaccin Immunother ; 15(11): 2778-2781, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31112447

RESUMO

Our laboratory has focused on Porin B (PorB), an outer membrane protein from Neisseria meningitidis and TLR2 ligand-based adjuvant, to characterize specific molecular and cellular pathways involved in improved immune responses induced by vaccine adjuvants. PorB's ability to form micellar nanoparticular multi-molecular organized structures and its interaction with Toll-like receptor 2/1 complexes likely accounts for its potent adjuvant activity. Downstream from this stimulation, we have observed enhanced antigen uptake in antigen presenting cells (APC), greater antigen deposition in secondary lymphoid organs, and promotion of germinal center reactions. In mice, antigen-specific IgGs were increased after PorB adjuvanted vaccination using the model antigen ovalbumin (OVA). Likewise, this formulation resulted in more IL-4 and IFN-γ positive T cells. Mice that received PorB adjuvanted vaccinations benefitted from lower bacterial burdens when challenged with recombinant Listeria monocytogenes expressing OVA. Mouse models lacking MyD88 signaling in various APC types helped identify macrophages as an essential cell type for the adjuvant activity of PorB. We believe the work presented here provides examples of the mechanistic studies required to understand how vaccine adjuvants are contributing to the establishment of protective immunity.


Assuntos
Adjuvantes Imunológicos , Células Apresentadoras de Antígenos/imunologia , Infecções Meningocócicas/prevenção & controle , Porinas/imunologia , Animais , Animais Geneticamente Modificados , Anticorpos Antibacterianos/sangue , Citocinas/imunologia , Feminino , Imunoglobulina G/sangue , Camundongos , Fator 88 de Diferenciação Mieloide , Neisseria meningitidis , Porinas/administração & dosagem , Linfócitos T/imunologia
2.
Bull Exp Biol Med ; 166(6): 751-753, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31028578

RESUMO

In vivo experiments showed that antibodies to OmpC and OmpF porins of Yersinia pseudotuberculosis increased thyroxine (T4) level in the blood of experimental animals. The mice were immunized with different antigens: recombinant OmpF porin in a soluble monomeric form, trimers of OmpC and OmpF porins isolated from the outer membrane, or antibodies to them. The level of thyroxine in the blood of mice immunized with OmpF and OmpC porins increased by 5.47 and 22.3 times, respectively; after immunization with antibodies to these proteins, blood thyroxine increased by 9.28 and 14.29 times. Immunization with recombinant OmpF porin induced no reliable increase in thyroxine level. Hence, the serum to recombinant OmpF porin contains no antibodies specific to conformational antigenic determinants that are present in the protein trimer and, according to our previous findings from molecular docking studies, determine cross-reactions between OmpF porin of Y. pseudotuberculosis and thyroidstimulating hormone receptor.


Assuntos
Antígenos de Bactérias/imunologia , Hipertireoidismo/induzido quimicamente , Porinas/imunologia , Yersinia pseudotuberculosis/química , Animais , Anticorpos Antibacterianos/administração & dosagem , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/química , Feminino , Hipertireoidismo/imunologia , Hipertireoidismo/metabolismo , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Porinas/administração & dosagem , Porinas/química , Multimerização Proteica , Receptores da Tireotropina/imunologia , Receptores da Tireotropina/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Tiroxina/biossíntese , Yersinia pseudotuberculosis/imunologia
3.
Vaccine ; 36(50): 7689-7699, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30381152

RESUMO

Vaccines formulated with adjuvant have been effective against numerous infectious diseases, almost always due to induction of functional antibodies that recognizes the pathogen of interest. There is an unmet clinical need for vaccine adjuvants that induce T cells responses to potentially enhance protection against malignancies and intracellular pathogens, where a humoral response, alone, may not be adequate for protection. In this study, we demonstrate that a TLR2 ligand-based adjuvant, meningococcal PorB, has broad immunostimulatory activity with the ability to induce a robust and diverse vaccine antigen specific T cell response. We demonstrate that a vaccine formulated with PorB admixed with ovalbumin induces a wide variety of antigen specific antibody subclasses and effector molecules (MIG, MCP-1, IP-10, MIP-1α, KC & IL-2) with known roles for inducing T cell responses, along with elevated levels of Th1 and Th2 type cytokines upon antigen stimulation. We confirmed production of these cytokines by examining the antigen-specific T cells induced by PorB in vivo. After two immunizations with vaccine formulated with PorB/OVA, antigen-specific CD4 and CD8 T cells were significantly increased in numbers and produced IL-4 or IFN-γ upon ex vivo antigen re-stimulation. Finally, in a Listeria mouse infection model, vaccine formulated with PorB significantly reduced the bacterial burden upon a low dose infection and increased survival upon a high dose infection with recombinant Listeria monocytogenes engineered to express OVA (rLmOVA), a pathogen that requires OVA-antigen specific cytotoxic CD8 T cells for clearance. In summary, PorB is able to induce antigen specific broad B and T cell responses, illustrating its potential as a potent and new vaccine adjuvant.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Imunidade Celular , Ovalbumina/imunologia , Porinas/administração & dosagem , Linfócitos T/imunologia , Animais , Carga Bacteriana , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Citocinas/análise , Modelos Animais de Doenças , Listeria monocytogenes/isolamento & purificação , Listeriose/patologia , Listeriose/prevenção & controle , Camundongos Endogâmicos C57BL , Ovalbumina/administração & dosagem , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
4.
Fish Shellfish Immunol ; 80: 563-572, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29958980

RESUMO

The outer-membrane proteins (OMPs) of Aeromonas hydrophila, an imperative fish pathogen accountable for massive economic losses to aquaculture industry, are found to be immunogenic and considered as potential vaccine candidates. In spite of development in the formulation of vaccine candidates against Aeromonas infection, no commercial preparation has been done so far; in addition, the molecular mechanisms of immunoprotection induced by various vaccine formulations in Indian major carp, Labeo rohita, are little known. The present study was undertaken to evaluate the modulation of immunity and expression of immune-related genes post-rOmpF (recombinant outer-membrane protein of A. hydrophila, a novel vaccine candidate) immunization and protective efficacy after A. hydrophila challenge. The rOmpF-immunized fish showed a variable expression of the immune-related genes, viz. toll-like receptor 22 (TLR), complement component 3 (C3), chemokine (CXCa), tumor necrosis factor-α (TNFα), interleukin 1ß (IL-1ß), manganese superoxide dismutase (MnSOD) and natural killer enhancing factor (NKEF) in the head kidney tissues, when compared to the control group at different time intervals post-vaccination. A significant increase in serum hemolysin titer, ceruloplasmin level and myeloperoxidase activity was observed on day 140 post immunization. Also, bacterial agglutination titer and antiprotease activity were significantly increased on day 42 post immunization. No significant change was observed in lysozyme activity. Challenge studies with live A. hydrophila on day 140 post-immunization of L. rohita significantly increased the relative percentage survival (∼44%) in the vaccinated group. The results suggest that the rOmpF could be used as a potential vaccine candidate to combat A. hydrophila infection in fish.


Assuntos
Aeromonas hydrophila/imunologia , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Vacinas Bacterianas/administração & dosagem , Cyprinidae/imunologia , Porinas/administração & dosagem , Animais , Anticorpos Antibacterianos/sangue , Proteínas da Membrana Bacteriana Externa/imunologia , Vacinas Bacterianas/imunologia , Ceruloplasmina/análise , Cyprinidae/sangue , Doenças dos Peixes/prevenção & controle , Infecções por Bactérias Gram-Negativas/prevenção & controle , Infecções por Bactérias Gram-Negativas/veterinária , Hemólise , Muramidase/sangue , Peroxidase/sangue , Porinas/imunologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia
5.
J Control Release ; 285: 12-22, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-29964134

RESUMO

Nanocarriers based on inverse hexagonal liquid crystalline phases (hexosomes) show promising potential as vaccine delivery systems. Their unique internal structure, composed of both lipophilic domains and water-containing channels, renders them capable of accommodating immunopotentiating compounds and antigens. However, their adjuvant properties are poorly understood. We hypothesized that the supramolecular structure of the lyotropic liquid crystalline phase influences the immunostimulatory activity of lipid-based nanocarriers. To test this, hexosomes were designed containing the lipid phytantriol (Phy) and the immunopotentiator monomycoloyl glycerol-1 (MMG-1). Self-assembly of Phy and MMG-1 into nanocarriers featuring an internal hexagonal phase was confirmed by small-angle X-ray scattering and cryogenic transmission electron microscopy. The effect of the nanostructure on the adjuvant activity was studied by comparing the immunogenicity of Phy/MMG-1 hexosomes with MMG-1-containing lamellar liquid crystalline nanoparticles (liposomes, CAF04). The quality and magnitude of the elicited immune responses were determined after vaccination of CB6/F1 mice using the Chlamydia trachomatis major outer membrane protein (MOMP) as antigen. MMG-1-based hexosomes potentiated significantly stronger MOMP-specific humoral responses than CAF04 liposomes. The liposome-based vaccine formulation induced a much stronger MOMP-specific cell-mediated immune response compared to hexosome-adjuvanted MOMP, which elicited minimal MOMP-specific T-cell stimulation after vaccination. Hence, our data demonstrates that hexosomal and liposomal adjuvants activate the immune system via different mechanisms. Our work provides valuable insights into the adjuvant potential of hexosomes and emphasizes that engineering of the supramolecular structure can be used to design adjuvants with customized immunological properties.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vacinas Bacterianas/farmacologia , Infecções por Chlamydia/prevenção & controle , Chlamydia trachomatis/imunologia , Álcoois Graxos/farmacologia , Monoglicerídeos/farmacologia , Porinas/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/química , Animais , Formação de Anticorpos/efeitos dos fármacos , Vacinas Bacterianas/administração & dosagem , Infecções por Chlamydia/imunologia , Portadores de Fármacos/química , Álcoois Graxos/administração & dosagem , Álcoois Graxos/química , Feminino , Cristais Líquidos/química , Camundongos , Monoglicerídeos/administração & dosagem , Monoglicerídeos/química , Nanopartículas/química , Porinas/administração & dosagem , Vacinação
6.
Sci Rep ; 6: 29353, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27383250

RESUMO

Common features of immune-metabolic and inflammatory diseases such as metabolic syndrome, diabetes, obesity and cardiovascular diseases are an altered gut microbiota composition and a systemic pro-inflammatory state. We demonstrate that active immunization against the outer membrane protein of bacteria present in the gut enhances local and systemic immune control via apoE-mediated immune-modulation. Reduction of western-diet-associated inflammation was obtained for more than eighteen weeks after immunization. Immunized mice had reduced serum cytokine levels, reduced insulin and fasting glucose concentrations; and gene expression in both liver and visceral adipose tissue confirmed a reduced inflammatory steady-state after immunization. Moreover, both gut and atherosclerotic plaques of immunized mice showed reduced inflammatory cells and an increased M2 macrophage fraction. These results suggest that adaptive responses directed against microbes present in our microbiota have systemic beneficial consequences and demonstrate the key role of apoE in this mechanism that could be exploited to treat immune-metabolic diseases.


Assuntos
Imunidade Adaptativa , Apolipoproteínas E/fisiologia , Aterosclerose/prevenção & controle , Dieta Ocidental , Microbioma Gastrointestinal/imunologia , Inflamação/prevenção & controle , Animais , Apolipoproteínas E/sangue , Proteínas de Bactérias/administração & dosagem , Glicemia/metabolismo , Citocinas/biossíntese , Citocinas/genética , Hormônios/sangue , Hormônios/genética , Insulina/sangue , Gordura Intra-Abdominal/metabolismo , Fígado/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Porinas/administração & dosagem
7.
Appl Biochem Biotechnol ; 178(4): 640-53, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26487188

RESUMO

The aims of this study were to identify alternative myxovirus (Mx) stimulatory compounds in Cirrhinus mrigala and to characterize the kinetics and intensity of their stimulated responses by semi-quantitative RT-PCR. Mx transcripts were measured in C. mrigala injected with Aeromonas OmpC (outer membrane protein) at a dose 0.4 mg/fish. At day 1, day 2, day 3, day 5, day 10, day 20 and day 30, samples were collected from kidney, spleen, liver, heart brain, gill, intestine and muscle for the study of Mx transcript and housekeeping gene ß-actin. Similarly, Mx gene expression was also studied in Aeromonas hydrophila-infected fish for a period of 10 days. Mx/ß-actin ratio was constitutively expressed from all the organs of OmpC-vaccinated fish. The expression was significantly highest (P ≤ 0.05) in spleen, followed by liver, kidney, intestine, gill, heart, muscle and brain. The expression was highest in day 2 except spleen (on day 3) and subsequently reduced up to day 30. Control fish also showed Mx expression. Similarly, A. hydrophila-infected fish showed Mx/ß-actin ratio upregulated significantly in the spleen and kidney on day 5, liver on day 2 and intestine on day 3. This study revealed that OmpC of A. hydrophila and its infection could stimulate the antiviral Mx gene of C. mrigala.


Assuntos
Aeromonas hydrophila/metabolismo , Cyprinidae/genética , Infecções por Bactérias Gram-Negativas/genética , Porinas/administração & dosagem , Animais , Eletroforese em Gel de Ágar , Cinética , RNA Mensageiro/genética
8.
Biotechnol Lett ; 38(4): 689-700, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26712371

RESUMO

OBJECTIVES: Involvement of the outer membrane protein C (OmpC) of Escherichia coli in neurodegeneration was investigated using a mouse model. RESULTS: OmpC formed protease-resistant fibres that exhibited the diagnostic features of an amyloid. The spectral shift in the Congo Red and the thioflavin T assays produced features similar to neurotoxic peptides. Intramuscular administration of OmpC in mice resulted in spongiform neurodegeneration of the brain through calcium-dependent apoptosis and also showed upregulation of apoptosis related genes. Immunolocalization of OmpC in brain demonstrated the direct involvement of the porin in neurodegeneration and formation of spongiform encephalopathy. CONCLUSION: We have demonstrated the ability of OmpC of E. coli to induce neurodegeneration in mice.


Assuntos
Encéfalo/patologia , Doenças Neurodegenerativas/etiologia , Porinas/toxicidade , Animais , Apoptose , Encéfalo/metabolismo , Cálcio/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Injeções Intramusculares , Camundongos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Porinas/administração & dosagem
9.
Hum Vaccin Immunother ; 11(3): 776-88, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25750999

RESUMO

The Neisseria meningitidis outer membrane protein PorA from a Chilean strain was purified as a recombinant protein. PorA mixed with AbISCO induced bactericidal antibodies against N. meningitidis in mice. When PorA was fused to the Helicobacter pylori HpaA antigen gene, the specific response against H. pylori protein increased. Splenocytes from PorA-immunized mice were stimulated with PorA, and an increase in the secretion of IL-4 was observed compared with that of IFN-γ. Moreover, in an immunoglobulin sub-typing analysis, a substantially higher IgG1 level was found compared with IgG2a levels, suggesting a Th2-type immune response. This study revealed a peculiar behavior of the purified recombinant PorA protein per se in the absence of AbISCO as an adjuvant. Therefore, the resistance of PorA to proteolytic enzymes, such as those in the gastrointestinal tract, was analyzed, because this is an important feature for an oral protein adjuvant. Finally, we found that PorA fused to the H. pylori HpaA antigen, when expressed in Lactococcus lactis and administered orally, could enhance the antibody response against the HpaA antigen approximately 3 fold. These observations strongly suggest that PorA behaves as an effective oral adjuvant.


Assuntos
Adesinas Bacterianas/imunologia , Adjuvantes Imunológicos/administração & dosagem , Anticorpos Antibacterianos/sangue , Helicobacter pylori/imunologia , Porinas/imunologia , Adesinas Bacterianas/administração & dosagem , Adesinas Bacterianas/genética , Adjuvantes Imunológicos/genética , Administração Oral , Animais , Feminino , Imunoglobulina G/sangue , Interferon gama/metabolismo , Interleucina-4/metabolismo , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Leucócitos Mononucleares/imunologia , Camundongos Endogâmicos BALB C , Porinas/administração & dosagem , Porinas/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Soro/química , Baço/imunologia
10.
PLoS One ; 8(10): e77084, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098576

RESUMO

BACKGROUND: The USA 2004 influenza virus outbreak H3N8 in dogs heralded the emergence of a new disease in this species. A new inactivated H3N8 vaccine was developed to control the spread of the disease but, as in humans and swine, it is anticipated that the virus will mutate shift and drift in the dog population. Therefore, there is a need for a vaccine that can trigger a broad protection to prevent the spread of the virus and the emergence of new strains. METHODOLOGY AND PRINCIPAL FINDINGS: The universal M2e peptide is identical in almost all the H3N8 influenza strains sequenced to date and known to infect dogs. This epitope is therefore a good choice for development of a vaccine to provide broad protection. Malva mosaic virus (MaMV) nanoparticles were chosen as a vaccine platform to improve the stability of the M2e peptide and increase its immunogenicity in animals. The addition of an adjuvant (OmpC) purified from Salmonella typhi membrane in the vaccine formulation increased the immune response directed to the M2e peptide significantly and enlarged the protection to include the heterosubtypic strain of influenza in a mouse model. An optimal vaccine formulation was also shown to be immunogenic in dogs. CONCLUSIONS AND SIGNIFICANCE: The MaMV vaccine platform triggered an improved immune response directed towards the universal M2e peptide. The adjuvant OmpC increased the immune response to the M2e peptide and protection to a heterosubtypic influenza strain that harbors a different M2e peptide in a mouse model. Antibodies generated by the vaccine formulation showed cross-reactivity with M2e peptides derived from influenza strains H9N2, H5N1 and H1N1. The vaccine formulation shows a potential for commercialization of a new M2e based vaccine in dogs.


Assuntos
Anticorpos Antivirais/sangue , Doenças do Cão/prevenção & controle , Vírus da Influenza A Subtipo H3N8/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/veterinária , Vacinação , Proteínas do Envelope Viral/imunologia , Adjuvantes Imunológicos/administração & dosagem , Sequência de Aminoácidos , Animais , Sequência Conservada , Proteção Cruzada , Doenças do Cão/imunologia , Doenças do Cão/virologia , Cães , Vírus da Influenza A Subtipo H1N1/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Vírus da Influenza A Subtipo H9N2/imunologia , Vacinas contra Influenza/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Vírus do Mosaico/genética , Vírus do Mosaico/imunologia , Nanopartículas/química , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Porinas/administração & dosagem , Proteínas do Envelope Viral/genética
11.
Immunology ; 139(4): 459-71, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23432484

RESUMO

Salmonella enterica serovar Typhi (S. Typhi) is the causal agent of typhoid fever, a disease that primarily affects developing countries. Various antigens from this bacterium have been reported to be targets of the immune response. Recently, the S. Typhi genome has been shown to encode two porins--OmpS1 and OmpS2--which are expressed at low levels under in vitro culture conditions. In this study, we demonstrate that immunizing mice with either OmpS1 or OmpS2 induced production of specific, long-term antibody titres and conferred protection against S. Typhi challenge; in particular, OmpS1 was more immunogenic and conferred greater protective effects than OmpS2. We also found that OmpS1 is a Toll-like receptor 4 (TLR4) agonist, whereas OmpS2 is a TLR2 and TLR4 agonist. Both porins induced the production of tumour necrosis factor and interleukin-6, and OmpS2 was also able to induce interleukin-10 production. Furthermore, OmpS1 induced the over-expression of MHC II molecules in dendritic cells and OmpS2 induced the over-expression of CD40 molecules in macrophages and dendritic cells. Co-immunization of OmpS1 or OmpS2 with ovalbumin (OVA) increased anti-OVA antibody titres, the duration and isotype diversity of the OVA-specific antibody response, and the proliferation of T lymphocytes. These porins also had adjuvant effects on the antibody response when co-immunized with either the Vi capsular antigen from S. Typhi or inactivated 2009 pandemic influenza A(H1N1) virus [A(H1N1)pdm09]. Taken together, the data indicate that OmpS1 and OmpS2, despite being expressed at low levels under in vitro culture conditions, are potent protective immunogens with intrinsic adjuvant properties.


Assuntos
Adjuvantes Imunológicos , Anticorpos Antibacterianos/sangue , Proteínas da Membrana Bacteriana Externa/imunologia , Porinas/imunologia , Vacinas contra Salmonella/imunologia , Salmonella typhi/imunologia , Febre Tifoide/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/genética , Animais , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Proteínas da Membrana Bacteriana Externa/genética , Células Dendríticas/imunologia , Relação Dose-Resposta a Droga , Feminino , Células HEK293 , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Imunização , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Ativação Linfocitária , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Ovalbumina/imunologia , Polissacarídeos Bacterianos/imunologia , Porinas/administração & dosagem , Porinas/genética , Vacinas contra Salmonella/administração & dosagem , Vacinas contra Salmonella/genética , Salmonella typhi/genética , Linfócitos T/imunologia , Fatores de Tempo , Receptor 2 Toll-Like/agonistas , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/metabolismo , Transfecção , Fator de Necrose Tumoral alfa/metabolismo , Febre Tifoide/sangue , Febre Tifoide/imunologia , Febre Tifoide/microbiologia
12.
Vaccine ; 31(7): 1065-71, 2013 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-23273968

RESUMO

BACKGROUND: An improved nonavalent PorA native outer membrane vesicle vaccine was developed with intrinsic adjuvating activity due to presence of less-toxic (lpxL1) LPS. In the present study, the safety and immunogenicity of this next-generation NonaMen vaccine were evaluated following repeated vaccination in rabbits and mice. METHODS: A repeated-dose toxicology study was performed in rabbits. Immunogenicity of next-generation NonaMen was evaluated by determining the serum bactericidal antibody (SBA) titers against meningococcal serogroup B strains containing several PorA subtypes. Release of the pro-inflammatory cytokine, interleukin-6 (IL-6), by the human monocytic cell line (MM6) was measured to estimate pyrogenic activity. RESULTS: No toxicologically relevant findings were noted in vaccinated rabbits receiving plain next-generation NonaMen. In agreement, next-generation NonaMen induced reduced amounts of the pro-inflammatory cytokine, IL-6, released by human monocyte cell line. In both rabbits and mice, next-generation NonaMen induced high SBA titers against all tested MenB strains regardless of whether or not aluminium phosphate adjuvant is used. CONCLUSIONS: The data suggest that next-generation NonaMen is a safe vaccine with the potential to develop a broadly protective immune response and encourage the start of the first clinical studies.


Assuntos
Antígenos de Bactérias/imunologia , Vacinas Meningocócicas/efeitos adversos , Vacinas Meningocócicas/imunologia , Neisseria meningitidis Sorogrupo B/imunologia , Porinas/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos , Animais , Antígenos de Bactérias/administração & dosagem , Atividade Bactericida do Sangue , Feminino , Interleucina-6/metabolismo , Masculino , Camundongos , Viabilidade Microbiana , Monócitos/imunologia , Porinas/efeitos adversos , Coelhos , Vacinação/efeitos adversos , Vacinação/métodos
13.
Methods Mol Biol ; 799: 319-41, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21993654

RESUMO

The process of cellular apoptosis is mediated by a number of microbial pathogens to modulate host defense mechanisms. Inhibition of apoptosis is thought to favor microbial survival, replication or immune evasion, while induction of apoptosis is likely to promote escape of the organisms from host cells. Several studies have reported that infection with Neisseria spp. can inhibit or reduce apoptotic cell death, thus allowing adaptation, intracellular replication, and immune evasion, events that are likely to spread infection. In this chapter, various techniques are described for direct measurement of host cell responses to infection with Neisseria meningitidis and to treatment with pure Neisseria porins, the major proteins found in the outer membrane of the pathogen.


Assuntos
Apoptose/fisiologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune/fisiologia , Neisseria meningitidis/fisiologia , Porinas/metabolismo , Western Blotting , Técnicas de Cultura de Células/métodos , Eletroforese em Gel de Poliacrilamida/métodos , Citometria de Fluxo , Células HeLa , Humanos , Indóis , Microscopia de Fluorescência , Porinas/administração & dosagem , Porinas/isolamento & purificação , Azul Tripano
14.
Vaccine ; 29(13): 2437-43, 2011 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-21276442

RESUMO

IC31(®), a novel adjuvant, has been shown to be effective by increasing the levels of IFN-γ in animal models when delivered with several antigens. Here, we tested the ability of IC31(®), to enhance the protective ability of the Chlamydia trachomatis native major outer membrane protein (nMOMP). BALB/c mice were immunized by the intramuscular (i.m.) and subcutaneous (s.c.) routes with nMOMP+IC31(®). Another group of animals was immunized with nMOMP+Alum and as a negative control mice were immunized with ovalbumin (Ova)+IC31(®). Animals immunized with nMOMP+IC31(®) developed high Chlamydia-specific IgG titers. The serum levels of IgG1 were higher than those of the IgG2a. T cells, from the spleens of mice immunized with IC31(®)-adjuvanted nMOMP demonstrated a strong lymphoproliferative reaction to Chlamydia elementary bodies (EB) compared with the groups immunized with nMOMP+Alum or Ova+IC31(®). A similar comparison between these groups of mice revealed that the levels of IFN-γ in the supernatants from stimulated T-cells were significantly higher in animals immunized with nMOMP+IC31(®). Following an intranasal challenge with C. trachomatis, the mice immunized with IC31(®)-adjuvanted nMOMP showed significant protection. The change in body weight, an indication of the severity of the infection, was significantly less reduced in mice immunized with nMOMP+IC31(®). Furthermore, the weight of the lungs, as well as the pulmonary Chlamydia burden, was significantly lower in the animals immunized with nMOMP+IC31(®) when compared with the groups immunized with nMOMP+Alum or with Ova+IC31(®). In conclusion, these results provide the rationale for further preclinical testing of IC31(®) using other chlamydial antigens, and support the potential evaluation of this adjuvant in human vaccines against Chlamydia.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas Bacterianas/imunologia , Infecções por Chlamydia/prevenção & controle , Oligodesoxirribonucleotídeos/administração & dosagem , Oligopeptídeos/administração & dosagem , Porinas/imunologia , Animais , Anticorpos Antibacterianos/sangue , Vacinas Bacterianas/administração & dosagem , Peso Corporal , Proliferação de Células , Chlamydia trachomatis/patogenicidade , Combinação de Medicamentos , Feminino , Imunoglobulina G/sangue , Injeções Intramusculares , Injeções Subcutâneas , Interferon gama/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Porinas/administração & dosagem , Índice de Gravidade de Doença , Baço/imunologia , Linfócitos T/imunologia
15.
Vaccine ; 29(7): 1399-407, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21195805

RESUMO

Plasmid DNA (pWRG7079::MOMP) expressing the major outer membrane protein of a human Chlamydia trachomatis serovar E strain was tested for the ability to induce an immune response and protect against experimental genital infection with the same serovar. The vaccine was tested in pigs, as they are genetically and physiologically related to humans and suitable for studying C. trachomatis infection of the genital system. To increase the immune response, GM-CSF, LTA and B and CpG motives were used as adjuvants. GM-CSF was administered seven days before immunization, while the other adjuvants were administered together with the vaccine. Ten pigs were randomly divided into two groups. One group received an intravaginal primo-vaccination and a booster of 500 µg pWRG7079::MOMP, while the other group received the placebo vaccine pWRG7079. All animals were challenged with 10(8) TCID(50) of C. trachomatis serovar E. Pigs immunized with the DNA vaccine showed significantly less macroscopic lesions, vaginal excretion and chlamydial replication in the genital tract, as compared to placebo-vaccinated controls. However, infection could not be completely cleared.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Vacinas Bacterianas/imunologia , Infecções por Chlamydia/prevenção & controle , Porinas/imunologia , Vagina/microbiologia , Adjuvantes Imunológicos/administração & dosagem , Administração Intravaginal , Animais , Anticorpos Antibacterianos/sangue , Especificidade de Anticorpos , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Derrame de Bactérias , Vacinas Bacterianas/administração & dosagem , Proliferação de Células , Infecções por Chlamydia/imunologia , Chlamydia trachomatis/imunologia , Citocinas/imunologia , Feminino , Leucócitos Mononucleares/imunologia , Porinas/administração & dosagem , Suínos , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Vagina/imunologia
16.
Artigo em Russo | MEDLINE | ID: mdl-20464999

RESUMO

AIM: To obtain recombinant atoxic form of Pseudomonas aeruginosa exotoxin A and assess its protective properties during simultaneous administration with recombinant protein F in experiment. MATERIALS AND METHODS: Genetic methods were employed for construction of deletion variant of P. aeruginosa exotoxin A gene, whereas Escherichia coli cells were used for transformation. Purification of proteins was performed by common method. White outbred mice were used for immunization and experimental infection was produced by intraperitoneal administration of live virulent culture of P. aeruginosa strain PA103. RESULTS: The gene coding defect form of P. aeruginosa exotoxin A with lacked 106 C-terminal aminoacid residues was cloned. Synthesized protein was nontoxic and immunogenic. Recombinant variants of anatoxin A and outer membrane protein F of P. aeruginosa protected animals from experimental infection caused by toxigenic strain PA103 of P. aeruginosa if were administered separately or concomitantly. Nonetheless, the highest protective effect was observed after immunization with both proteins. CONCLUSION: Studied recombinant toxoid and OprF could be the candidates for inclusion in vaccines for prevention of pseudomonas infection.


Assuntos
ADP Ribose Transferases/imunologia , Toxinas Bacterianas/imunologia , Exotoxinas/imunologia , Porinas/imunologia , Infecções por Pseudomonas/prevenção & controle , Vacinas contra Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Proteínas Recombinantes/imunologia , Fatores de Virulência/imunologia , ADP Ribose Transferases/administração & dosagem , Animais , Toxinas Bacterianas/administração & dosagem , Exotoxinas/administração & dosagem , Injeções Intraperitoneais , Camundongos , Porinas/administração & dosagem , Vacinas contra Pseudomonas/administração & dosagem , Pseudomonas aeruginosa/química , Proteínas Recombinantes/administração & dosagem , Vacinação , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Fatores de Virulência/administração & dosagem , Exotoxina A de Pseudomonas aeruginosa
17.
Microbes Infect ; 10(6): 657-63, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18462971

RESUMO

Several in vivo models have been used to dissect the molecular mechanisms that contribute to activate the coagulation and fibrinolytic systems by bacteria and bacterial products but many aspects remain poorly understood. In this study we examined the in vivo effect of the synthetic peptide corresponding to loop L7 from Haemophilus influenzae type b (Hib) porin to evaluate its role on the coagulative/fibrinolytic cascade and the circulating markers of endothelial injury. Plasma was obtained from rats injected intravenously with loop L7, Hib porin or a scrambled peptide and tested for fragment 1+2 (F1+2), tissue-type plasminogen activator (tPA), plasminogen activator inhibitor type I (PAI-1) antigen, von Willebrand factor (vWF) and soluble E-selectin (sE-selectin). The coagulative/fibrinolytic cascade was impaired as shown by PAI-1 level increased. Concomitantly, E-selectin, a marker of endothelial injury, was also significantly elevated. In addition either loop L7 or Hib porin injection induced hyperglycaemia and inflammatory cytokine production. The data were correlated with hemodynamic functions. The results indicate that loop L7 plays an essential role in the pathophysiologic events observed during gram-negative infection. These findings may have implications for the development of alternative therapies to counteract excessive inflammatory responses during septic shock.


Assuntos
Proteínas de Bactérias/administração & dosagem , Infecções por Bactérias Gram-Negativas/fisiopatologia , Haemophilus influenzae/química , Porinas/administração & dosagem , Motivos de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Fibrinólise , Infecções por Bactérias Gram-Negativas/metabolismo , Peptídeos/administração & dosagem , Peptídeos/síntese química , Peptídeos/química , Porinas/síntese química , Ratos , Ratos Sprague-Dawley
18.
Vaccine ; 26(6): 786-96, 2008 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-18191311

RESUMO

Porins from pathogenic Neisseriae are among several bacterial products with immune adjuvant activity. Neisseria meningitidis (Nme) PorB, has been shown to induce immune cells activation in a TLR2-dependent manner and acts as a vaccine immune adjuvant. The PorB porin from Neisseria lactamica (Nlac), a common nasopharyngeal commensal, shares significant structural and functional similarities with Nme PorB. In this work we ask whether the immune adjuvant ability of porins from pathogenic Neisserial strains is a characteristic shared with porins from non-pathogenic Neisserial species or whether it is unique for bacterial products derived from microorganisms capable of inducing inflammation and disease. We evaluated the potential immune adjuvant effect of Nlac PorB in mice using ovalbumin (OVA) as a prototype antigen. Immunization with Nlac PorB/OVA induced high OVA-specific IgG and IgM titers compared to OVA alone, similar to other adjuvants such as Nme PorB and alum. High titers of IgG1 and IgG2b were detected as well as production of IL-4, IL-10, IL-12 and INF-gamma in response to Nlac PorB, consistent with induction of both a Th1-type and a Th2-type immune response. OVA-specific proliferation was also determined in splenocytes from Nlac PorB/OVA-immunized mice. In addition, B cell activation in vitro and cytokine production in response to Nlac PorB was found to be mediated by TLR2, in a similar manner to Nme PorB.


Assuntos
Adjuvantes Imunológicos , Anticorpos/sangue , Imunização , Neisseria lactamica/imunologia , Ovalbumina/imunologia , Porinas/imunologia , Células Th1/imunologia , Células Th2/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos/imunologia , Especificidade de Anticorpos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Células Cultivadas , Esquemas de Imunização , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Injeções Subcutâneas , Interferon gama/biossíntese , Interleucinas/biossíntese , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/administração & dosagem , Porinas/administração & dosagem , Baço/imunologia , Células Th1/metabolismo , Células Th2/metabolismo
19.
Appl Environ Microbiol ; 74(3): 833-9, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18065632

RESUMO

Campylobacter species are important enteric pathogens causing disease in humans and animals. There is a lack of a good immunological test that can be used routinely to separate Campylobacter jejuni from other Campylobacter species. We produced monoclonal antibodies (MAbs) directed against the major outer membrane protein (MOMP) of C. jejuni using recombinant MOMP as the antigen. One MAb, designated MAb5C4 and of the immunoglobulin G1 isotype, was found to be potentially specific for C. jejuni. Dot blots demonstrated that MAb5C4 reacted with all 29 isolates of C. jejuni tested but did not react with 2 C. jejuni isolates, 26 other Campylobacter spp. isolates, and 19 non-Campylobacter isolates. Western blotting showed that MAb5C4 bound to a single protein band approximately 43 kDa in size, corresponding to the expected size of C. jejuni MOMP. The detection limit of MAb5C4 in a dot blot assay was determined to be about 5 x 10(3) bacteria. The epitope on the MOMP was mapped to a region six amino acids in length with the sequence 216GGQFNP221, which is 97% conserved among C. jejuni strains but divergent in other Campylobacter spp.; a GenBank search indicated that 95% of C. jejuni isolates will be able to be detected from non-Campylobacter spp. based on the highly specific and conserved region of the GGQFNP polypeptide. The epitope is predicted to be located in a region that is exposed to the periplasm. MAb5C4 is a potentially specific and sensitive MAb that can be used for the specific detection and identification of C. jejuni.


Assuntos
Anticorpos Monoclonais/biossíntese , Especificidade de Anticorpos , Proteínas de Bactérias/imunologia , Campylobacter jejuni/imunologia , Epitopos/imunologia , Porinas/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Mapeamento de Epitopos , Epitopos/química , Humanos , Hibridomas , Imunização , Immunoblotting , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Porinas/administração & dosagem , Porinas/química , Porinas/genética , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Análise de Sequência de DNA
20.
Int Immunol ; 20(1): 81-8, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18003602

RESUMO

Macrophages treated with porin of Shigella dysenteriae were potent stimulators of naive T(h) cells since the porin-pulsed macrophages strongly proliferated the T cells and up-regulated the activation molecules CD69 and CD25 on CD4(+) T cells in allogeneic mixed leukocyte reaction. Immunization of C57BL/6 mice with porin selectively induced the intracellular expression and release of IFN-gamma and had no effect on IL-4 expression. In parallel to the predominant release of the T(h)1 cytokine IFN-gamma, up-regulation of CCR5 on the immune CD4(+) T cells and messenger ribonucleic acid (mRNA) expression of the T cell chemokines macrophage inflammatory protein-1 alpha and regulated on activation normal T cell expressed and secreted showed T(h)1 bias. The porin-primed CD4(+) T cells expressed the mRNA for T-box expressed in T cells, a T(h)1-specific transcription factor confirming the adjuvant-induced transition of T cells to polarized effector T(h)1 cells. The immune CD4(+) T cells proliferated and released IL-2 and IFN-gamma profoundly in response to re-challenge with porin-pulsed macrophages but not to BSA-pulsed macrophages in vitro, which demonstrated the presence of porin-specific CD4(+) T cells of T(h)1 phenotype. The study highlights that porin has the capacity of an adjuvant to unfold and maintain the T cell function and thereby to activate adaptive immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Ativação Linfocitária/imunologia , Porinas/imunologia , Shigella dysenteriae/imunologia , Células Th1/imunologia , Animais , Técnicas de Cocultura , Humanos , Imunização , Interferon gama/metabolismo , Teste de Cultura Mista de Linfócitos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Porinas/administração & dosagem , Shigella dysenteriae/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA