Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
ACS Appl Mater Interfaces ; 13(13): 14964-14973, 2021 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-33769023

RESUMO

The low efficacy of current conventional treatments for bacterial infections increases mortality rates worldwide. To alleviate this global health problem, we propose drug-free enzyme-based nanomotors for the treatment of bacterial urinary-tract infections. We develop nanomotors consisting of mesoporous silica nanoparticles (MSNPs) that were functionalized with either urease (U-MSNPs), lysozyme (L-MSNPs), or urease and lysozyme (M-MSNPs), and use them against nonpathogenic planktonic Escherichia coli. U-MSNPs exhibited the highest bactericidal activity due to biocatalysis of urea into NaHCO3 and NH3, which also propels U-MSNPs. In addition, U-MSNPs in concentrations above 200 µg/mL were capable of successfully reducing 60% of the biofilm biomass of a uropathogenic E. coli strain. This study thus provides a proof-of-concept, demonstrating that enzyme-based nanomotors are capable of fighting infectious diseases. This approach could potentially be extended to other kinds of diseases by selecting appropriate biomolecules.


Assuntos
Antibacterianos/farmacologia , Escherichia coli/efeitos dos fármacos , Muramidase/farmacologia , Nanopartículas/química , Dióxido de Silício/química , Urease/farmacologia , Antibacterianos/administração & dosagem , Biocatálise , Biofilmes/efeitos dos fármacos , Canavalia/enzimologia , Portadores de Fármacos/química , Escherichia coli/fisiologia , Infecções por Escherichia coli/tratamento farmacológico , Humanos , Muramidase/administração & dosagem , Urease/administração & dosagem , Infecções Urinárias/tratamento farmacológico
2.
Front Immunol ; 9: 1003, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29867978

RESUMO

Vaccination is an increasingly important alternative approach to control Helicobacter pylori infection, since H. pylori resistance to previously efficacious antibiotic regimens is increased, and H. pylori eradication treatment for upper gastrointestinal diseases is becoming less successful. Fortunately, an efficient oral monovalent H. pylori vaccine has been developed. However, compared with monovalent vaccines, multivalent vaccines have the potential to induce more effective and comprehensive protection against H. pylori infection. In this study, we designed and produced a multivalent epitope-based vaccine cholera toxin B subunit (CTB)-HUUC with the intramucosal adjuvant CTB and tandem copies of B-cell epitopes (HpaA132-141, UreA183-203, and UreB321-339) and T-cell epitopes (HpaA88-100, UreA27-53, UreB229-251, UreB317-329, UreB373-385, UreB438-452, UreB546-561, CagA149-164, and CagA196-217) from H. pylori adhesion A subunit (HpaA), urease A subunit (UreA), urease B subunit (UreB), and cytotoxin-associated antigen (CagA). Serum IgG, stomach, and intestine mucosal sIgA from mice after CTB-HUUC vaccination neutralized H. pylori urease activity in vitro. CTB-HUUC vaccination promoted H. pylori-specific lymphocyte responses and a mixed CD4+ T cell immune response as indicated by IFN-γ, interleukin-4, and interleukin-17 production in mice. Both oral prophylactic and therapeutic CTB-HUUC vaccinations reduced gastric urease activity and H. pylori infection and protected stomachs in mice. Taken together, CTB-HUUC is a promising potent and safe multivalent vaccine in controlling H. pylori infection in BALB/c mouse model.


Assuntos
Toxina da Cólera/imunologia , Vacinas contra Cólera/imunologia , Infecções por Helicobacter/prevenção & controle , Urease/imunologia , Administração Oral , Animais , Anticorpos Antibacterianos/sangue , Toxina da Cólera/administração & dosagem , Vacinas contra Cólera/administração & dosagem , Modelos Animais de Doenças , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito T/imunologia , Feminino , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Urease/administração & dosagem , Vacinação , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
3.
Sci Rep ; 5: 14793, 2015 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-26434384

RESUMO

Helicobacter pylori (H. pylori) infects more than half of the world's population, causing chronic gastritis, peptic ulcers and gastric cancer. Urease B subunit (UreB), a conserved protein of H. pylori, is capable of inducing specific CD4(+) T-cell responses and provides protection against this infection. Previous studies have confirmed the effectiveness of rUreB subunit vaccines in generating CD4(+) T-cell-mediated protection, but less is known regarding the roles of different subtypes of T-cell immunity, such as Th1, Th2 and Th17, particularly the immunodominant epitopes inducing specific CD4(+) T-cell responses, in vaccine-mediated protection. In this study, we demonstrated that the vaccination of BALB/c mice with rUreB resulted in significant antigen-specific Th1 and Th17 immune responses. Importantly, two novel Th epitopes, UreB317-329 and UreB409-421, which are recognized by a major population of CD4(+) T cells, were identified in immunized mice. Our results demonstrated that two novel epitopes can simultaneously induce Th1 and Th17 immune responses; however, only the epitope vaccine-induced CD4(+) T-cells secreting IFN-γ mediated the protection against H. pylori; cells secreting IL-17A did not. Taken together, our results suggest that two novel immunodominant epitopes can induce Th1 and Th17 immune responses, but only the induced Th1 lymphocytes mediate protection against H. pylori.


Assuntos
Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Células Th1/fisiologia , Células Th17/fisiologia , Vacinação , Adjuvantes Imunológicos/administração & dosagem , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/química , Proteínas de Bactérias/imunologia , Células Cultivadas , Mapeamento de Epitopos , Epitopos de Linfócito T/imunologia , Feminino , Adjuvante de Freund/administração & dosagem , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Interferon gama/metabolismo , Interleucina-17/metabolismo , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Células Th1/microbiologia , Células Th17/microbiologia , Urease/administração & dosagem , Urease/química , Urease/imunologia
4.
Biochem Biophys Res Commun ; 462(3): 269-74, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-25957472

RESUMO

Urease is considered as an excellent vaccine candidate antigen against Helicobacter pylori (H. pylori) infection. Our previous study reported a novel multi-epitope vaccine CTB-UE which was composed of the mucosal adjuvant cholera toxin B subunit (CTB) and five cell epitopes from urease subunits. Murine experiments indicated that it could induce cellular and humoral immune responses intensively and attenuate H. pylori infection effectively in mice model. However, the body expression and lack of suitable adjuvant of this epitope vaccine restricted its application. In this study, new recombinant Escherichia coli strains was established to increase the solubility by fusing thioredoxin (Trx) and the combination adjuvants which composed of the chitosan and CpG were adopted to enhance the immunogenicity of CTB-UE for oral immunization. The experimental results indicated that the levels of IgG2a, IgG1 and IgA in the serum and the levels of sIgA in stomach, intestine and feces were significantly higher in the vaccinated group compared with the model control group. Additionally, chitosan-CpG combination adjuvants changed the ratio of IgG2a/IgG1 and conferred Th1/Th17-mediated protective immune responses. These results demonstrate that the oral vaccine with chitosan-CpG as combination adjuvants may be a promising vaccine candidate against H. pylori infection.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Quitosana/administração & dosagem , Quitosana/imunologia , Toxina da Cólera/administração & dosagem , Toxina da Cólera/imunologia , Ilhas de CpG/imunologia , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Urease/administração & dosagem , Urease/imunologia , Administração Oral , Animais , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/sangue , Especificidade de Anticorpos , Vacinas Bacterianas/genética , Citocinas/biossíntese , Epitopos/administração & dosagem , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/enzimologia , Helicobacter pylori/genética , Imunidade nas Mucosas , Imunoglobulina A Secretora/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Urease/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
5.
J Comp Pathol ; 148(2-3): 266-77, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22901429

RESUMO

Pulmonary sarcoidosis, a human disease of unknown cause, has no animal model. Sarcoidosis patients have serum antibodies specific for Helicobacter pylori and its surface enzyme urease. H. pylori do not survive in the high-oxygen pulmonary atmosphere, but urease may access the lung by oesophageal reflux. A model was established in rats to study gastro-oesophageal reflux of urease into the airways. Pathology in tissues from human sarcoidosis patients was compared with that in the rat model. Changes observed in the rat model included prominent peribronchial lymphocytic infiltration, which is seen occasionally in human sarcoidosis. Granulomas, pathognomonic for human sarcoidosis, occurred occasionally in the lungs of rats given urease protein intratracheally, but were widespread when urease was coupled to microbeads and administered intravenously. Biomarkers associated with human sarcoidosis (interleukin1-ß and platelet-activating factor) were up-regulated acutely in the rat model. Further investigations with this model may provide significant insights into the origin and pathogenesis of pulmonary diseases in man and other species that carry gastric Helicobacter spp. and its associated enzyme.


Assuntos
Antígenos de Bactérias/sangue , Modelos Animais de Doenças , Granuloma/imunologia , Helicobacter pylori/imunologia , Pneumopatias/imunologia , Sarcoidose Pulmonar/imunologia , Urease/imunologia , Administração Oral , Adulto , Animais , Feminino , Refluxo Gastroesofágico/complicações , Granuloma/metabolismo , Granuloma/patologia , Humanos , Interleucina-1beta/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Pneumopatias/metabolismo , Pneumopatias/patologia , Linfócitos/patologia , Masculino , Pessoa de Meia-Idade , Fator de Ativação de Plaquetas/metabolismo , Ratos , Ratos Sprague-Dawley , Sarcoidose Pulmonar/metabolismo , Sarcoidose Pulmonar/patologia , Urease/administração & dosagem
6.
Appl Microbiol Biotechnol ; 95(6): 1437-44, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22569640

RESUMO

Epitope vaccine based on the enzyme urease of Helicobacter pylori is a promising option for prophylactic and therapeutic vaccination against H. pylori infection. In our previous study, the epitope vaccine CTB-UA, which was composed of the mucosal adjuvant cholera toxin B subunit (CTB) and an epitope (UreA183₋203) from the H. pylori urease A subunit (UreA) was constructed. This particular vaccine was shown to have good immunogenicity and immunoreactivity and could induce specific neutralizing antibodies, which exhibited effectively inhibitory effects on the enzymatic activity of H. pylori urease. In this study, the prophylactic and therapeutic efficacy of the epitope vaccine CTB-UA was evaluated in a BALB/c mice model. The experimental results indicated that oral prophylactic or therapeutic immunization with CTB-UA significantly decreased H. pylori colonization compared with oral immunization with PBS. The results also revealed that the protection was correlated with antigen-specific IgG, IgA, and mucosal secretory IgA antibody responses. CTB-UA may be a promising vaccine candidate for the control of H. pylori infection.


Assuntos
Proteínas de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Toxina da Cólera/imunologia , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Urease/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/genética , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/genética , Toxina da Cólera/administração & dosagem , Toxina da Cólera/genética , Modelos Animais de Doenças , Infecções por Helicobacter/tratamento farmacológico , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/enzimologia , Helicobacter pylori/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Urease/administração & dosagem , Urease/genética , Vacinação
8.
Gastroenterology ; 141(4): 1273-82, 1282.e1, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21703999

RESUMO

BACKGROUND & AIMS: Protective immunization limits Helicobacter infection of mice by undetermined mechanisms. Protease-activated receptor 2 (PAR2) signaling is believed to regulate immune and inflammatory responses. We investigated the role of PAR2 in vaccine-induced immunity against Helicobacter infection. METHODS: Immune responses against Helicobacter infection were compared between vaccinated PAR2-/- and wild-type (WT) mice. Bacterial persistence, gastric pathology, and inflammatory and cellular responses were assessed using the rapid urease test (RUT), histologic analyses, quantitative polymerase chain reaction, and flow cytometry, respectively. RESULTS: Following vaccination, PAR2-/- mice did not have reductions in Helicobacter felis infection (RUT values were 0.01±0.01 for WT mice and 0.11±0.13 for PAR2-/- mice; P<.05). The vaccinated PAR2-/- mice had reduced inflammation-induced stomach tissue damage (tissue damage scores were 8.83±1.47 for WT mice and 4.86±1.35 for PAR2-/- mice; P<.002) and reduced T-helper (Th)17 responses, based on reduced urease-induced interleukin (IL)-17 secretion by stomach mononuclear cells (5182 ± 1265 pg/mL for WT mice and 350±436 pg/mL for PAR2-/- mice; P<.03) and reduced recruitment of CD4+ IL-17+ T cells into the gastric mucosa of PAR2-/- mice following bacterial challenge (3.7%±1.5% for WT mice and 2.6%±1.1% for PAR2-/- mice; P<.05). In vitro, H felis-stimulated dendritic cells (DCs) from WT mice induced greater secretion of IL-17 by ovalbumin-stimulated OT-II transgenic CD4+ T cells compared with DCs from PAR2-/- mice (4298±347 and 3230±779; P<.04), indicating that PAR2-/- DCs are impaired in priming of Th17 cells. Adoptive transfer of PAR2+/+ DCs into vaccinated PAR2-/- mice increased vaccine-induced protection (RUT values were 0.11±0.10 and 0.26±0.15 for injected and noninjected mice, respectively; P<.03). CONCLUSIONS: PAR2 activates DCs to mediate vaccine-induced protection against Helicobacter infection in mice.


Assuntos
Vacinas Bacterianas/administração & dosagem , Infecções por Helicobacter/prevenção & controle , Helicobacter felis/imunologia , Helicobacter pylori/imunologia , Receptor PAR-2/metabolismo , Estômago/efeitos dos fármacos , Urease/administração & dosagem , Administração Intranasal , Transferência Adotiva , Animais , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Células Dendríticas/transplante , Modelos Animais de Doenças , Feminino , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/enzimologia , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor PAR-2/deficiência , Receptor PAR-2/genética , Baço/efeitos dos fármacos , Baço/imunologia , Baço/microbiologia , Estômago/imunologia , Estômago/microbiologia , Estômago/patologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/microbiologia , Vacinas Sintéticas/administração & dosagem
9.
Artigo em Inglês | MEDLINE | ID: mdl-19900066

RESUMO

Kidney failure is a common disease with high frequency. Food-grade recombinant bacteria that can effectively remove urea has great potential for treatment of renal failure. A nonpathogenic strain, L. lactis MG1363, was transformed with plasmid pMG36eure, which carries urease gene. The expression of transgene urease in genetically modified L. lactis MG1363 and the urease activity in removal of urea were investigated. It was found that the removal of urea by recombinant L. lactis MG1363 was pH- and nickel-dependent. At pH 6.5 and in the presence of 250 microM of NiSO4, 50 approximately 60% of urea could be removed in 24 hours. The urea removal activity was also evaluated in imitative gastroenteric environment. After being exposed to acidic solution (pH2.5-4.0) for 2 hours, the cells were then grown in a medium containing 0.1 cfu/ml bile acid salt, 30 mg/dl urea, and 250 microM NiSO4 at pH 6.8. The concentration of urea decreased over time, and the removal was about 30% at 10 hours and 65% at 24 hours, respectively. The safety tests were performed by feeding normal rats with either L. lactis MG1363 or recombinant L. lactis MG1363. The two materials did not cause any changes in blood cells and blood biochemical indexes. There were no differences in terms of body weight and water/food consumption between the two materials. These results indicate the safety, feasibility, and capacity of urease gene modified Lactococcus Lactis in removal of urea under the gastroenteric circumstances. Further investigation may generate a food-grade strain for treatment of chronic renal failure.


Assuntos
Terapia Biológica/métodos , Lactococcus lactis/genética , Ureia/metabolismo , Urease/metabolismo , Animais , Engenharia Genética , Concentração de Íons de Hidrogênio , Nefropatias/terapia , Níquel/farmacologia , Ratos , Proteínas Recombinantes , Transformação Bacteriana , Transgenes , Resultado do Tratamento , Urease/administração & dosagem , Urease/genética , Urease/uso terapêutico
10.
J Microencapsul ; 25(8): 569-76, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18465293

RESUMO

In this paper a device, based on urease-loaded microspheres, is presented. The first task of this work was the optimization of a procedure for the alginate microspheres realization, having a radius as close as possible to the optimal one necessary to achieve the maximum enzyme exploitation. This optimal radius was calculated theoretically through a mathematical model which describes the concentration of substrate (urea) inside the microspheres on the assumption of a diffusion-reaction mechanism. The enzyme-loaded microspheres were successfully tested in a prototypal device aimed at the depletion of urea from a circulating fluid simulating blood flow: the results showed that urea concentration in the circulating fluid drops down to less than 25% of the initial value after 5 h.


Assuntos
Alginatos/química , Portadores de Fármacos/química , Microesferas , Ureia/metabolismo , Urease/uso terapêutico , Alginatos/uso terapêutico , Enzimas Imobilizadas/administração & dosagem , Enzimas Imobilizadas/metabolismo , Enzimas Imobilizadas/uso terapêutico , Ácido Glucurônico/química , Ácido Glucurônico/uso terapêutico , Ácidos Hexurônicos/química , Ácidos Hexurônicos/uso terapêutico , Humanos , Nefropatias/terapia , Tamanho da Partícula , Ureia/sangue , Urease/administração & dosagem , Urease/metabolismo
11.
Int J Pharm ; 349(1-2): 226-33, 2008 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-17825506

RESUMO

In this study, the effects of alginate modification on absorption properties of FITC-BSA loaded TMC nanoparticles were investigated on an in vitro model of GI epithelium (Caco-2 cells). The feasibility of applying TMC nanoparticles loaded with a model vaccine urease in oral vaccination was also studied. Alginate modified TMC nanoparticles showed higher FITC-BSA permeate efficiency than non-modified TMC nanoparticles. However, alginate modification barely had any effect on TMC nanoparticles' property of decreasing TEER or enhancing drug paracellular transport. Mice s.c. immunized with urease loaded TMC nanoparticles showed highest systematic immune response (IgG levels) but the lowest mucosal response (secretory IgA levels). In the contrast, mice i.g. immunized with urease loaded TMC nanoparticles showed much higher antibody titers of both IgG and secretory IgA than those with urease solution or urease co-administrated with TMC solution. These results indicated that TMC nanoparticles are potential carriers for oral protein and vaccine delivery.


Assuntos
Quitosana/química , Proteínas/administração & dosagem , Administração Oral , Alginatos , Animais , Vacinas Bacterianas/administração & dosagem , Células CACO-2 , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Eletroquímica , Ensaio de Imunoadsorção Enzimática , Feminino , Fluoresceína-5-Isotiocianato , Corantes Fluorescentes , Helicobacter pylori/imunologia , Humanos , Imunização , Camundongos , Nanopartículas , Tamanho da Partícula , Soroalbumina Bovina/administração & dosagem , Soroalbumina Bovina/farmacocinética , Urease/administração & dosagem , Urease/imunologia
12.
Arch Biochem Biophys ; 452(2): 149-55, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16839515

RESUMO

Ureases (EC 3.5.1.5) are highly homologous enzymes found in plants, bacteria and fungi. Canatoxin, an isoform Canavalia ensiformis urease, has several biological properties unrelated to its ureolytic activity, like platelet-aggregating and pro-inflammatory effects. Here, we describe that Bacillus pasteurii urease (BPU) also induces aggregation of rabbit platelets, similar to the canatoxin-induced effect (ED(50) 0.4 and 0.015 mg/mL, respectively). BPU induced-aggregation was blocked in platelets pretreated with dexamethasone and esculetin, a phospholipase A(2) and a lipoxygenase inhibitor, respectively, while platelets treated with indomethacin, a cyclooxygenase inhibitor, showed increased response to BPU. Methoxyverapamil (Ca(2+) channel blocker) and AMP (ADP antagonist) abrogated urease-induced aggregation, whereas the PAF-acether antagonist Web2170 had no effect. We concluded that platelet aggregation induced by BPU is mediated by lipoxygenase-derived eicosanoids and secretion of ADP from the platelets through a calcium-dependent mechanism. Potential relevance of these findings for bacterium-plant interactions and pathogenesis of bacterial infections are discussed.


Assuntos
Bacillus/enzimologia , Plaquetas/fisiologia , Fabaceae/enzimologia , Proteínas de Plantas/administração & dosagem , Agregação Plaquetária/fisiologia , Urease/administração & dosagem , Animais , Plaquetas/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Agregação Plaquetária/efeitos dos fármacos , Coelhos
13.
Vaccine ; 24(18): 3832-9, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16122847

RESUMO

Vaccination against Helicobacter pylori is of particular clinical interest. Recombinant urease, the major protein in H. pylori, has been used for mucosal vaccination trials in different animal models, but was found to be ineffective in humans. The current study therefore investigated the human immune response towards recombinant H. pylori urease A and B (rUreA/B) expressed in E. coli compared to different cellular fractions of H. pylori (cytosol, total, inner and outer membrane). Monocyte-derived dendritic cells (Mo-DC) were generated from monocytes isolated by magnetic antigen cell separation (MACS) from healthy volunteers and cultured in the presence of hrIL-4 and hrGM-CSF. Mo-DC were stimulated for 48h with the recombinant proteins (1 microg/ml) or cellular fractions (1-10 microg/ml) and cytokine release was determined in the culture supernatant by ELISA. rUreA and rUreB were effective in inducing IL-12 secretion (6-10 fold) and, to a much lesser extent (2 fold), IL-10 secretion from Mo-DC. Total and outer membrane preparations from H. pylori stimulated IL-12 secretion significantly, and were even more potent than intact bacteria. Mo-DCs pulsed with rUreA activated allogenic CD56+ NK-cells, as determined by TNF-alpha and IFN-gamma secretion, but not allogenic CD4+/CD45RA+ naïve T-cells. In contrast, Mo-DCs pulsed with H. pylori total membrane or outer membrane preparations activated allogenic naive T-cells in co-culture systems, as determined by increased TNF-alpha secretion. It appears that outer membrane preparations of H. pylori, but not recombinant urease are more effective in inducing a Th1 polarized response in humans in vitro.


Assuntos
Vacinas Bacterianas/imunologia , Membrana Celular/imunologia , Citosol/imunologia , Helicobacter pylori/imunologia , Linfócitos T/imunologia , Urease/imunologia , Vacinas Sintéticas/imunologia , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/genética , Células Cultivadas , Citocinas/biossíntese , Células Dendríticas/imunologia , Ensaio de Imunoadsorção Enzimática , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Helicobacter pylori/enzimologia , Helicobacter pylori/genética , Humanos , Interferon gama/biossíntese , Interleucina-10/análise , Interleucina-12/análise , Interleucina-4/imunologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Células Th1/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Urease/administração & dosagem , Urease/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética
14.
J Infect Dis ; 192(8): 1441-9, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16170763

RESUMO

BACKGROUND: The development of an efficacious vaccine against infection with Helicobacter pylori, the causative agent of chronic gastritis, peptic ulcer disease, and gastric adenocarcinoma, remains a challenge. Since the use of mucosal adjuvants is limited in human application, we have evaluated the potential of recombinant Lactobacillus strains producing H. pylori urease B (UreB) subunit to deliver this antigen to the gastrointestinal tract. METHODS: Mice were injected orally 3 times with a triple dose of recombinant Lactobacillus plantarum NCIMB8826, the recombinant isogenic cell-wall mutant (alr(-) MD007 strain) expressing UreB, or a mixture of recombinant UreB and cholera toxin (rUreB/CT) as a control. Urease-specific seric immunoglobulin (Ig) G and IgA were measured by use of an enzyme-linked immunosorbent assay. After challenge with Helicobacter felis, stomach infection was examined by use of the rapid urease test and by polymerase chain reaction detection of Helicobacter genomic DNA. RESULTS: Intragastric immunization with both recombinant Lactobacillus strains and rUreB/CT elicited UreB-specific antibodies. After challenge, reduction of H. felis load in the stomachs of mice was observed only after immunization with the recombinant mutant strain MD007 or with rUreB/CT. CONCLUSIONS: This is the first report of successful induction of partial protection against H. felis with a mucosal prime-boost regimen in which recombinant Lactobacillus strains were used as antigen-delivery vehicles.


Assuntos
Vacinas Bacterianas/administração & dosagem , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/enzimologia , Imunoglobulina A Secretora/biossíntese , Lactobacillus/imunologia , Urease/administração & dosagem , Adjuvantes Imunológicos , Administração Oral , Animais , Formação de Anticorpos , Vacinas Bacterianas/imunologia , Modelos Animais de Doenças , Helicobacter/isolamento & purificação , Infecções por Helicobacter/imunologia , Helicobacter pylori/genética , Imunoglobulina A Secretora/sangue , Imunoglobulina G/biossíntese , Imunoglobulina G/sangue , Lactobacillus/genética , Camundongos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Urease/biossíntese , Urease/genética , Urease/imunologia
15.
Acta Biochim Pol ; 52(1): 261-6, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15827623

RESUMO

Following genetic immunization of laying ducks with a plasmid expressing Helicobacter pylori UreB (large subunit of urease), IgY against UreB were obtained from egg yolks. These polyclonal and monospecific IgY antibodies are of higher-titer and specifically recognize recombinant H. pylori urease purified from Escherichia coli. To our knowledge this is the first report describing generation of IgY antibodies directed against antigens of H. pylori by DNA-based immunization.


Assuntos
Anticorpos Antibacterianos/biossíntese , Patos/imunologia , Gema de Ovo/imunologia , Helicobacter pylori/imunologia , Urease/administração & dosagem , Animais , Anticorpos Antibacterianos/imunologia , Ensaio de Imunoadsorção Enzimática , Helicobacter pylori/enzimologia , Imunoglobulinas/imunologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Urease/genética , Urease/imunologia
16.
Vaccine ; 23(7): 901-9, 2005 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-15603891

RESUMO

We developed a novel vaccine for Helicobacter pylori based on a poliovirus vector in which capsid genes were replaced with the gene for the B subunit of H. pylori urease (UreB). Mice were vaccinated with UreB or control (L1) replicon and challenged with H. pylori. Twenty percent of mice vaccinated prophylactically with UreB, but 80% vaccinated with L1, and then challenged with H. pylori became infected (P = 0.003). Seventy-three percent of mice with established H. pylori infection vaccinated therapeutically with UreB replicon cleared their infection compared to 33% vaccinated with L1 (P = 0.067). In therapeutically vaccinated mice with residual infection, UreB-vaccinated animals had fewer H. pylori than L1-vaccinated mice (P < 0.05). Anti-urease antibody titres in prophylactically, but not therapeutically, vaccinated mice were markedly higher in animals that received UreB versus L1 replicon (P = 0.01). Vaccination with poliovirus vector containing the gene for the B subunit of H. pylori urease provides significant prophylactic and strong therapeutic protection against H. pylori in mice.


Assuntos
Vacinas Bacterianas/administração & dosagem , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/enzimologia , Helicobacter pylori/genética , Poliovirus/genética , Replicon/genética , Urease/genética , Animais , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Células HeLa , Infecções por Helicobacter/genética , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Poliovirus/imunologia , Replicon/imunologia , Urease/administração & dosagem , Urease/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia
17.
J Dairy Sci ; 87(12): 4073-9, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15545368

RESUMO

Helicobacter pylori is a human pathogen that infects over 50% of the population worldwide. It is the most important etiologic agent of gastroduodenal ulcers and malignancies. Helicobacter pylori urease enzyme is considered the main factor for the organism's colonization in the gastroduodenal mucosa. Hens immunized with the purified urease produce a highly specific anti-H. pylori urease immunoglobulin (IgY-urease) in their egg yolks. Immunoglobulin Y-urease was stable at 60 to 65 degrees C for 30 min and at pH 4.0 for 7 h. Its activity was lost at 80 degrees C for 20 min and at pH 2 for 4 h. Specially designed functional drinking yogurt containing Lactobacillus acidophilus and Bifidobacterium spp. with 1% egg yolk IgY-urease was produced commercially. Immunoglobulin Y-urease activity showed stability in the product up to 7 d, and then decreased to 85% after 3 wk of storage. A clinical study was conducted to determine the effectiveness of IgY-urease yogurt to suppress infection in humans. Forty-two volunteers who tested positive for H. pylori using a 13C-urea breath test were recruited. A total of 450 mL of IgY-urease (test group) or IgY-urease-free yogurt (control group) was consumed in 150-mL portions 3 times daily for 4 wk. Volunteers were tested after 2 and 4 wk; urea breath test values significantly decreased in the test group compared with the control group. The results indicate that suppression of H. pylori infection in humans could be achieved by consumption of drinking yogurt fortified with IgY-urease.


Assuntos
Infecções por Helicobacter/dietoterapia , Helicobacter pylori , Imunoglobulinas/uso terapêutico , Urease/imunologia , Iogurte , Adulto , Animais , Testes Respiratórios , Galinhas , Gema de Ovo/imunologia , Feminino , Alimentos Orgânicos , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Humanos , Concentração de Íons de Hidrogênio , Terapia de Imunossupressão/métodos , Masculino , Pessoa de Meia-Idade , Temperatura , Urease/administração & dosagem
18.
Vaccine ; 21(3-4): 194-201, 2002 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-12450694

RESUMO

Low dose E. coli heat-labile enterotoxin (LT), delivered orally or enterically, has been used as an adjuvant for Helicobacter pylori (H. pylori) urease in healthy adults. In this study we aim to test the safety and adjuvant efficacy of LT delivered rectally together with recombinant H. pylori urease. Eighteen healthy adults without present or past H. pylori infection were enrolled in a double blind, randomized, ascending dose study to receive either urease (60 mg), or urease (60 mg) + LT (5 or 25 microg). The immunization preparation was administered per rectum on days 0, 14 and 28. Serum, stool and saliva anti-urease and anti-LT IgG and IgA antibodies (Abs) were measured and urease-specific and LT-specific antigen secreting cells (ASCs) were counted in peripheral blood at baseline and 7 (ASC counts) or 14 days (antibody levels) after each dosing. Peripheral blood lymphoproliferation assays were also performed at baseline and at the end of the study. Rectally delivered urease and LT were well tolerated. Among the 12 subjects assigned to urease+LT, 2 (16.7%) developed anti-urease IgG Abs, 1 (8.3%) developed anti-urease IgA Abs, and 3 (25%) showed urease-specific IgA(+) ASCs. Immune responses to LT were more vigorous, especially in subjects exposed to 5 microg LT. In the urease+ 5 microg LT group, anti-LT IgG and IgA Abs developed in 60 and 80% of the subjects, respectively, while LT-specific IgG(+) and IgA(+) ASCs were detected in all subjects. The magnitude of the anti-LT response was much higher than the response to urease. No IgA anti-urease or anti-LT Abs were detected in stool or saliva and lymphocyte proliferative responses to urease were unsatisfactory. In conclusion, rectal delivery of 5 microg LT is safe and induces vigorous systemic anti-LT immune responses. Further studies are needed to determine if LT can be an effective adjuvant for rectally delivered antigens.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Enterotoxinas/administração & dosagem , Vacinas contra Escherichia coli/imunologia , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/enzimologia , Urease/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos , Administração Retal , Anticorpos Antibacterianos/sangue , Enterotoxinas/efeitos adversos , Ensaio de Imunoadsorção Enzimática , Escherichia coli/química , Vacinas contra Escherichia coli/administração & dosagem , Vacinas contra Escherichia coli/efeitos adversos , Humanos , Imunização , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Segurança , Urease/imunologia , Vacinação
19.
Gut ; 51(5): 634-40, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12377799

RESUMO

BACKGROUND AND AIMS: Escherichia coli heat labile enterotoxin (LT) at doses of 5 micro g or 10 micro g has adjuvant activity for oral immunisation in humans infected with Helicobacter pylori, but causes severe diarrhoea. This study was undertaken to establish a safe and effective dose of LT, to confirm the safety of recombinant urease, and to compare the immunogenicity of orally compared with enterically delivered urease. METHODS: 42 healthy adults without present or past H pylori infection were randomised to receive 60 mg recombinant H pylori urease in soluble or in encapsulated form, given with doses of LT ranging from 0 micro g to 2.5 micro g. Four oral doses were administered at day 1, 8, 29, and 57. Specific IgG, IgA, and antibody secreting cells were measured as well as total alpha4beta7 integrin positive lymphocyte responses. RESULTS: Enterically delivered urease was well tolerated and no serious adverse events occurred. Mild diarrhoea (one to four loose stools) occurred after the first immunisation in 50% (6 of 12) of the volunteers exposed to 2.5 micro g LT (p=0.06; paired t test, compared with baseline) but not in volunteers exposed to lower LT doses. Immune responses occurred in five (p=0.048; Fisher's exact test), one, two, and one of six subjects exposed to 2.5 micro g, 0.5 micro g, 0.1 micro g, and no LT, respectively. Significant CD4(+), CD69(+), and CD45RO(hi) responses occurred over time among alpha4beta7(hi) lymphocytes in volunteers receiving 2.5 micro g LT. Enterically delivered urease induced higher lymphocyte responses than soluble urease. CONCLUSIONS: The safety of H pylori urease is confirmed. Oral LT may conserve its adjuvant activity at low doses with minimal side effects.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Toxinas Bacterianas/administração & dosagem , Enterotoxinas/administração & dosagem , Proteínas de Escherichia coli , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/enzimologia , Urease/administração & dosagem , Administração Oral , Adulto , Análise de Variância , Preparações de Ação Retardada , Diarreia/etiologia , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estatísticas não Paramétricas , Linfócitos T/imunologia , Urease/imunologia
20.
Scand J Immunol ; 53(5): 437-42, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11309150

RESUMO

Several vaccination studies have been performed in monkeys and humans testing the feasibility of prophylactic and therapeutic immunizations against Helicobacter pylori. The monkey studies showed that immune responses were induced by oral vaccination with the mucosal adjuvant LT (Escherichia coli heat-labile enterotoxin), parenteral administration with a cationic lipid adjuvant, and by mucosal priming followed by parenteral boosts. Both prophylactic and therapeutic activities were demonstrated in monkeys, providing a strong impetus for human vaccine trials. Preliminary studies in humans were undertaken in order to identify a tolerable dose of LT adjuvant or to test the effectiveness of mutant atoxic LT adjuvants. The results from these preliminary studies suggest that native LT causes diarrhoea at doses required for adjuvanticity while a mutant LT does not. In one study in which infected human subjects were vaccinated with orally administered urease antigen with native LT, there was a modest reduction in the level of H. pylori gastric colonization. A second clinical study employing H. pylori whole cell antigen and a mutant LT in infected subjects showed immune responses and although the subjects remained infected, the study was not designed to measure reduction in H. pylori colonization. Recombinant Salmonella expressing urease and other H. pylori antigens have been effective in mice (see accompanying Frontlines Topic Review by John O. Nedrud [1]), but monkey studies are not possible because of host range restriction. Human trials of parenteral immunization, mucosal immunization with mutant LT and live Salmonella vectors are needed to fully assess the ability of vaccines to prevent or treat H. pylori infections.


Assuntos
Vacinas Bacterianas/administração & dosagem , Modelos Animais de Doenças , Proteínas de Escherichia coli , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos , Animais , Anticorpos Antibacterianos/biossíntese , Antígenos de Bactérias/administração & dosagem , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/efeitos adversos , Ensaios Clínicos como Assunto , Enterotoxinas/administração & dosagem , Enterotoxinas/efeitos adversos , Previsões , Gastroenteropatias/prevenção & controle , Gastroenteropatias/terapia , Haplorrinos , Infecções por Helicobacter/terapia , Humanos , Urease/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA