Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Clin Exp Immunol ; 153(3): 456-62, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18637102

RESUMO

Immunoglobulin A (IgA) glycosylation, recognized as an important pathogenic factor in IgA nephropathy (IgAN), is apparently controlled by the polarity of T helper (Th) cytokine responses. To examine the role of cytokine polarity in IgAN, inbred mice were immunized by intraperitoneal priming with inactivated Sendai virus (SeV) emulsified in either complete Freund's adjuvant (CFA) or incomplete Freund's adjuvant (IFA), which promote Th1- or Th2-immune response, respectively, and then boosted identically twice orally with aqueous suspensions of inactivated virus. Next, some mice were challenged intranasally with infectious SeV. Mice primed with CFA or IFA had equal reductions in nasal viral titre relative to non-immune controls, and equally increased serum levels of SeV-specific IgA antibody. Mice primed with CFA showed higher SeV-specific IgG than those with IFA. Splenocytes from mice primed with IFA produced copious amounts of interleukin (IL)-4 and IL-5, but little interferon-gamma and IL-2; those primed with CFA had reciprocal cytokine recall responses. Total serum IgA and especially SeV-specific IgA from mice primed with IFA showed a selective defect in sialylation and galactosylation. Although the frequency and intensity of glomerular deposits and haematuria did not differ, glomerulonephritis in mice primed with IFA and challenged with infectious virus was more severe than in those given CFA, as judged by serum creatinine level. We conclude that the polarity of T cell cytokines controls the pattern of IgA glycosylation and exerts direct or indirect effects on functional glomerular responses to immune complex deposition.


Assuntos
Citocinas/imunologia , Glomerulonefrite por IGA/imunologia , Imunoglobulina A/imunologia , Vírus Sendai/imunologia , Células Th1/imunologia , Células Th2/imunologia , Adjuvantes Imunológicos , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Adjuvante de Freund , Glicosilação , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Lipídeos , Camundongos , Camundongos Endogâmicos BALB C , Líquido da Lavagem Nasal/imunologia , Células Th1/metabolismo , Células Th2/metabolismo
2.
Curr Top Microbiol Immunol ; 241: 181-213, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10087662

RESUMO

Studies in both humans and animals demonstrate that H. pylori is capable of illiciting an innate response that in part is regulated by the genetic makeup of the host. These innate responses includes stimulating immune effector mechanisms at the cellular and biochemical level resulting in the influx of neutrophils into the lamina propria and have even been shown to modify gastric acid secretion. The availability of good animal models of chronic Helicobacter infection has also allowed investigators to begin to examine how the adaptive host immune response prevents and/or exacerbates Helicobacter-induced gastroduodenal disease. The experimental H. felis/mouse model has been utilized by a number of laboratories to investigate mechanisms of host defense against chronic Helicobacter infection. This model and the more recently developed H. pylori rodent model has not only allowed investigators to confirm the feasibility of immunotherapy to prevent and/or cure Helicobacter infection but also to begin to examine how the host immune response prevents and/or exacerbates Helicobacter-induced gastroduodenal disease. Based on these studies a hypothesis is emerging that suggests that protection and/or cure from Helicobacter infection is mediated primarily by an upregulated cellular immune response which may act via an antibody independent mechanism. Paradoxically, following natural infection with H. pylori, a component of the cellular immune response also promotes chronic gastric inflammation without clearance of the organism. The recent development of reliable and reproducible H. pylori/rodent models of disease and the availability of numerous inbred strains, transgenic and knockout animals, will allow investigators to continue to explore the role the host cellular and humoral immune response plays in promoting or preventing this infection.


Assuntos
Vacinas Bacterianas/imunologia , Gastroenteropatias/imunologia , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Adjuvantes Imunológicos/administração & dosagem , Administração Oral , Animais , Anticorpos Antibacterianos/imunologia , Especificidade de Anticorpos , Vacinas Bacterianas/administração & dosagem , Ensaios Clínicos como Assunto , Citocinas/metabolismo , Modelos Animais de Doenças , Ácido Gástrico/metabolismo , Mucosa Gástrica/imunologia , Gastroenteropatias/microbiologia , Gastroenteropatias/terapia , Predisposição Genética para Doença , Infecções por Helicobacter/terapia , Humanos , Imunidade Ativa , Imunidade Celular , Imunidade Inata , Imunidade nas Mucosas/imunologia , Camundongos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
3.
Mucosal Immunol ; 8(5): 1047-59, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25603827

RESUMO

Lymphoid tissue inducer (LTi) cells are activated by accessory cell IL-23, and promote lymphoid tissue genesis and antibacterial peptide production by the mucosal epithelium. We investigated the role of LTi cells in the gastric mucosa in the context of microbial infection. Mice deficient in IRAK-M, a negative regulator of TLR signaling, were investigated for increased LTi cell activity, and antibody mediated LTi cell depletion was used to analyze LTi cell dependent antimicrobial activity. H. pylori infected IRAK-M deficient mice developed increased gastric IL-17 and lymphoid follicles compared to wild type mice. LTi cells were present in naive and infected mice, with increased numbers in IRAK-M deficient mice by two weeks. Helicobacter and Candida infection of LTi cell depleted rag1(-/-) mice demonstrated LTi-dependent increases in calprotectin but not RegIII proteins. However, pathogen and commensal microbiota populations remained unchanged in the presence or absence of LTi cell function. These data demonstrate LTi cells are present in the stomach and promote lymphoid follicle formation in response to infection, but are limited by IRAK-M expression. Additionally, LTi cell mediated antimicrobial peptide production at the gastric epithelium is less efficacious at protecting against microbial pathogens than has been reported for other tissues.


Assuntos
Candida/imunologia , Candidíase/imunologia , Mucosa Gástrica/imunologia , Infecções por Helicobacter/imunologia , Helicobacter/imunologia , Quinases Associadas a Receptores de Interleucina-1/imunologia , Tecido Linfoide/imunologia , Animais , Candidíase/genética , Candidíase/patologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Infecções por Helicobacter/genética , Infecções por Helicobacter/patologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Quinases Associadas a Receptores de Interleucina-1/genética , Interleucina-23/genética , Interleucina-23/imunologia , Tecido Linfoide/patologia , Camundongos , Camundongos Knockout
4.
Aliment Pharmacol Ther ; 12 Suppl 1: 83-90, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9701006

RESUMO

A large body of literature supports a role for Helicobacter pylori as an environmental factor which contributes to the development of gastric cancer through an intermediate stage of atrophic gastritis. The mechanism(s) by which H. pylori-associated chronic inflammation progresses to more serious diseases such as peptic ulcer and gastric cancer in certain individuals is not clear. Variations in the phenotype or genotype of the infecting H. pylori strain can play a role in the severity of disease. However, individuals infected with the 'more virulent' strains of H. pylori often never develop serious disease. Our experiments in inbred strains of mice provide evidence that host genetics also play a significant role in H. pylori-related gastritis. We have demonstrated that gastritis is dominated by a TH1 adaptive immune response, the degree of which directly correlates with the severity of disease. Treatment of mice with IL-12 or anti-IFN-gamma antibodies can increase or decrease, respectively the severity of gastritis in infected mice and adoptive transfer of TH1 cell lines significantly exacerbates disease. Thus, the tendency of an individual to respond to infection with specific immune mechanisms can dramatically affect the severity of disease and possibly put an individual at increased risk of progressing to disorders such as gastric cancer.


Assuntos
Gastrite/etiologia , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Neoplasias Gástricas/imunologia , Animais , Gastrite/patologia , Gastrite/terapia , Infecções por Helicobacter/complicações , Infecções por Helicobacter/genética , Helicobacter pylori/genética , Humanos , Interferon gama/imunologia , Interferon gama/uso terapêutico , Interleucina-12/imunologia , Interleucina-12/uso terapêutico , Camundongos , Fenótipo , Neoplasias Gástricas/etiologia
5.
Eur J Gastroenterol Hepatol ; 7 Suppl 1: S1-6, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8574728

RESUMO

OBJECTIVE: To examine the level and duration of the humoral immune response to Helicobacter felis following oral immunization or infection. DESIGN AND METHODS: Germ-free mice were orally immunized with sonicated H. felis plus cholera toxin five times over 6 weeks. One week after immunization was completed, immunized and control non-immunized mice received an oral challenge of live H. felis organisms. The animals were killed at 3-week intervals and serum, gastric washings, intestinal washings and gastric biopsies were obtained. H. felis infection was confirmed by a positive urease test or culture of the gastric biopsy. Serum gastric and intestinal antibody titers were determined by enzyme-linked immunosorbent assay. CONCLUSION: Infection and immunization against H. felis produces a specific humoral response. The humoral response in infection alone is significantly smaller than that of immunized animals until 6 weeks after infection. The humoral response following oral immunization persists for at least 18 weeks without further stimulation. The presence of an H. felis-specific antibody immune response before infection may be needed to protect animals from acute Helicobacter infection.


Assuntos
Anticorpos Antibacterianos/metabolismo , Vacinas Bacterianas/administração & dosagem , Toxina da Cólera/administração & dosagem , Sistema Digestório/imunologia , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori , Imunização Passiva , Administração Oral , Animais , Anticorpos Antibacterianos/sangue , Biópsia , Sistema Digestório/enzimologia , Ensaio de Imunoadsorção Enzimática , Vida Livre de Germes , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/patologia , Imunoglobulina A/administração & dosagem , Imunoglobulina G/administração & dosagem , Camundongos
6.
Can J Gastroenterol ; 13(7): 591-4, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10519957

RESUMO

Immunization can prevent or cure an otherwise chronic helicobacter infection in several animal models despite the chronic nature of natural helicobacter infections. Differences in the antigenic specificity of the antibodies may contribute to the protection observed in these experimental animals. The goal of the present study was to compare the local and systemic antibody responses of humans with chronic Helicobacter pylori infection with those of an individual with spontaneous resolution of infection to find an immunological correlate of protection. Spontaneous resolution of infection was accompanied by a change in immunoblot profiles. Whereas a broad range of H pylori antigens was recognized in chronically infected patients (including the patient who ultimately cleared the infection spontaneously), resolution of infection in the absence of therapeutic agents resulted in the recognition of only several immunodominant antigens. The most dominant antigen was approximately 66 kDa in molecular mass. Immunoblot analysis demonstrated that these antibodies were specific for the structural subunits of the urease enzyme. These studies suggest that the success of antihelicobacter immunization may be due to the ability of vaccination to induce an immune response against antigens that are normally not immunodominant during the course of infection.


Assuntos
Anticorpos Antibacterianos/imunologia , Gastroenteropatias/microbiologia , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Epitopos , Feminino , Humanos , Immunoblotting , Imunoglobulina A/imunologia , Masculino
7.
Curr Protoc Immunol ; Chapter 19: Unit 19.8, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-18432759

RESUMO

In this unit, protocols for growing Helicobacter organisms on plates or in liquid cultures are presented, followed by protocols for infecting mice with Helicobacter felis and H. pylori and for infecting ferrets with H. mustelae. Also, a procedure is described for adapting an H. pylori isolate to growth in mice. Support protocols describe methods for quantifying numbers of Helicobacter organisms, and how to create a growth curve for Helicobacter cultures. One important technique in investigating Helicobacter infection is assaying the disease processes that occur in the stomach, and a protocol is provided for preparing tissue sections for this purpose. It is also important to confirm that organisms recovered from tissue samples are, in fact, Helicobacter species, and a support protocol describes morphological and biochemical tests for this purpose. Helicobacter bacteria produce large amounts of the enzyme urease, and a support protocol describes how to perform a rapid urease test on animal-tissue biopsies. Assays of Helicobacter-specific immune responses require appropriate antigens, and preparation of both Helicobacter lysates and outer-membrane proteins are detailed for use in these assays.


Assuntos
Infecções por Helicobacter , Helicobacter , Modelos Animais , Adaptação Fisiológica , Animais , Biópsia , Furões , Mucosa Gástrica/metabolismo , Helicobacter/crescimento & desenvolvimento , Helicobacter/metabolismo , Infecções por Helicobacter/diagnóstico , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/fisiologia , Camundongos , Estômago/patologia , Urease/análise , Urease/metabolismo
8.
Gastroenterol Clin North Am ; 29(3): 671-85, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11030080

RESUMO

Since the initial discovery of H. pylori by Marshall and Warren 17 years ago, much progress has been made in treating this infection. However, as we enter the millennium, H. pylori infection continues to be one of the most common infections of mankind. In addition, eradication of H. pylori still requires multiple antimicrobial agents. A better understanding of the host immune response to H. pylori infection should allow investigators to develop immunotherapies to prevent the acquisition of infection and eradicate existing chronic H. pylori infection.


Assuntos
Vacinas Bacterianas/farmacologia , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Animais , Infecções por Helicobacter/imunologia , Humanos , Camundongos , Prevenção Primária/métodos , Sensibilidade e Especificidade
9.
Infect Immun ; 63(3): 1113-5, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7868235

RESUMO

Immunodeficient (SCID) and immunocompetent mice were infected with Helicobacter felis to address the role of autoimmunity in Helicobacter-associated gastritis. The extents of inflammation were equivalent in the two groups. The numbers of H. felis organisms were marginally increased in the SCID mice but did not achieve statistical significance. These results indicate that autoimmunity is not necessary to induce disease and that the presence of an adaptive immune system does not significantly affect H. felis colonization.


Assuntos
Gastrite/imunologia , Infecções por Helicobacter/imunologia , Estômago/patologia , Animais , Anticorpos Antibacterianos/sangue , Autoimunidade , Doença Crônica , Ensaio de Imunoadsorção Enzimática , Camundongos , Camundongos SCID , Estômago/microbiologia
10.
Immunology ; 94(1): 22-7, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9708182

RESUMO

Cholera toxin is a potent oral mucosal adjuvant for enteric immunization. Several studies suggest that commercial cholera toxin B subunit (cCTB; purified from holotoxin) may be an effective non-toxic alternative for oral immunization. The present study was performed, using an infectious disease model, to determine if the oral mucosal adjuvanticity of CTB is dependent on contaminating holotoxin. Mice were orally immunized with Helicobacter felis sonicate and either cholera holotoxin, cCTB or recombinant cholera toxin B subunit (rCTB). Serum immunoglobulin G (IgG) and intestinal immunoglobulin A (IgA) antibody responses were determined and the mice were challenged with live H. felis to determine the degree of protective immunity induced. All orally immunized mice responded with serum IgG antibody titres regardless of the adjuvant used. However, only mice immunized with either holotoxin or the cCTB responded with an intestinal mucosal IgA response. Consistent with the production of mucosal antibodies, mice immunized with either holotoxin or cCTB as adjuvants were protected from challenge while mice receiving H. felis sonicate and rCTB all became infected. cCTB induced the accumulation of cAMP in mouse thymocytes at a level equal to 0.1% of that induced by holotoxin, whereas rCTB was devoid of any activity. These results indicate that CTB possesses no intrinsic mucosal adjuvant activity when administered orally. Therefore, when used as an oral adjuvant, CTB should also include small, non-toxic doses of cholera toxin.


Assuntos
Adjuvantes Imunológicos , Toxina da Cólera/imunologia , Infecções por Helicobacter/prevenção & controle , Imunização , Administração Oral , Animais , Anticorpos Antibacterianos/biossíntese , Antígenos de Bactérias/imunologia , AMP Cíclico/biossíntese , Feminino , Helicobacter/imunologia , Imunidade nas Mucosas , Imunoglobulina A/biossíntese , Imunoglobulina G/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/imunologia
11.
J Infect Dis ; 179(3): 725-8, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9952387

RESUMO

Immunization can prevent or cure an otherwise chronic gastric Helicobacter infection in several different animal models. The goal of the present study was to compare the titers and specificities of local and systemic antibody responses generated by Helicobacter infection and immunization. Protective immunization results in levels of specific gastric antibody significantly lower than induced by infection. However, antibodies from protectively immunized mice preferentially recognize immunodominant proteins of 10-22 and 30 kDa. Immunoblot analysis of infected mice and humans demonstrated that the serum IgA, but not serum IgG, binding profiles yield an accurate profile of the antigenic specificity of the host's gastric IgA. Therefore, serum IgA may be useful in evaluating the immunodominant antigens at the gastric mucosa of infected persons and possibly in determining the immunogenicity of orally applied Helicobacter vaccines.


Assuntos
Vacinas Bacterianas , Infecções por Helicobacter/imunologia , Helicobacter pylori , Helicobacter/imunologia , Adulto , Idoso , Animais , Anticorpos Antibacterianos/imunologia , Formação de Anticorpos , Antígenos de Bactérias/imunologia , Dispepsia/imunologia , Dispepsia/microbiologia , Feminino , Mucosa Gástrica/imunologia , Helicobacter pylori/imunologia , Humanos , Imunoglobulina A/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Úlcera Péptica/imunologia , Úlcera Péptica/microbiologia
12.
J Immunol ; 166(6): 3688-92, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11238608

RESUMO

The capacity of mucosal IgA Abs to serve as an excretory immune system in vivo was investigated. Mice expressing a transgenic TCR were immunized intragastrically with the cognate Ag to elicit a vigorous mucosal IgA Ab response. Soon after i.v. challenge, Ag was detected within the epithelial cells of the small intestinal crypts and to a lesser degree within the epithelial cells higher up the villi, paralleling the gradient in expression of the polymeric Ig receptor and the transport of its ligand, oligomeric IgA. Uptake of Ag into the epithelial cells occurred only from the basolateral aspect and only when Ag complexed to IgA Ab could be present in the lamina propria. The results support the concept that local IgA Abs can excrete Ags from the body by transporting them directly through mucosal epithelial cells, using the same mechanism that transports free IgA into the mucosal secretions.


Assuntos
Imunoglobulina A/fisiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Animais , Especificidade de Anticorpos , Complexo Antígeno-Anticorpo/metabolismo , Biotinilação , Galinhas , Feminino , Imunidade nas Mucosas , Imunização Secundária , Imunoglobulina A/biossíntese , Imunoglobulina A/metabolismo , Injeções Intravenosas , Mucosa Intestinal/citologia , Intubação Gastrointestinal , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Ovalbumina/metabolismo
13.
Infect Immun ; 69(6): 4168-73, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11349094

RESUMO

The contribution of glutamyl transpeptidase (GGT) (gamma-glutamyltransferase [EC 2. 3. 2. 2]) to Helicobacter pylori virulence was investigated in piglets and mice using GGT-deficient isogenic strains. All animals became colonized. However, the bacterial load was significantly lower for mutant bacteria than for parent strains. These results suggest that GGT activity provides an advantage to H. pylori in colonization.


Assuntos
Infecções por Helicobacter/microbiologia , Helicobacter pylori/crescimento & desenvolvimento , Helicobacter pylori/patogenicidade , gama-Glutamiltransferase/metabolismo , Animais , Modelos Animais de Doenças , Mucosa Gástrica/microbiologia , Vida Livre de Germes , Helicobacter pylori/enzimologia , Helicobacter pylori/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Suínos , Virulência , gama-Glutamiltransferase/genética
14.
Helicobacter ; 1(1): 43-51, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9398912

RESUMO

BACKGROUND: Numerous clinical trials evaluating the efficacy of various antimicrobial compounds against Helicobacter pylori infection have been performed in humans. A convenient animal model for Helicobacter infection would facilitate the evaluation of novel therapies. These experiments were performed to evaluate the use of ferrets as a model of Helicobacter infection. MATERIALS AND METHODS: Ferrets were infected experimentally with Helicobacter mustelae and subsequently treated with bismuth subsalicylate (BSS) triple therapy (BSS, metronidazole, and amoxicillin), or left untreated. The status of infection and serology was assessed during treatment and for 8 weeks posttreatment. Seven ferrets successfully treated with triple therapy were challenged with H. mustelae and monitored for infection for an additional 5 weeks. RESULTS: Infection of ferrets by H. mustelae was accompanied by gastritis and a specific antibody response. Treatment of H. mustelae-infected ferrets with BSS suppressed bacterial growth in four of nine animals but did not eradicate infection. Triple therapy eradicated infection in all nine ferrets with a reduction in gastric inflammation. No relapse of infection occurred up to 8 weeks posttherapy. Challenge with H. mustelae of ferrets successfully treated with triple therapy resulted in a 100% rate of reinfection. CONCLUSIONS: H. mustelae infection can be eliminated by triple therapy, but this does not result in protective immunity against reinfection by H. mustelae. This model, using a strain of Helicobacter indigenous to the host, may be useful for assessing therapeutic efficacy of novel therapies for the treatment of human infection by H. pylori.


Assuntos
Amoxicilina/uso terapêutico , Antiácidos/uso terapêutico , Bismuto/uso terapêutico , Quimioterapia Combinada/uso terapêutico , Furões/microbiologia , Gastrite/tratamento farmacológico , Infecções por Helicobacter/tratamento farmacológico , Metronidazol/uso terapêutico , Compostos Organometálicos/uso terapêutico , Salicilatos/uso terapêutico , Amoxicilina/administração & dosagem , Animais , Antiácidos/administração & dosagem , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/imunologia , Bismuto/administração & dosagem , Quimioterapia Combinada/administração & dosagem , Feminino , Gastrite/imunologia , Gastrite/patologia , Helicobacter/imunologia , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/patologia , Masculino , Metronidazol/administração & dosagem , Compostos Organometálicos/administração & dosagem , Salicilatos/administração & dosagem , Organismos Livres de Patógenos Específicos
15.
Infect Immun ; 63(4): 1394-9, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7890401

RESUMO

Experiments were performed to determine the antigenic specificity of a monoclonal antibody (immunoglobulin A [IgA] 71) previously demonstrated to neutralize the ability of Helicobacter felis to colonize mice. Immunoprecipitation of radiolabeled H. felis outer membrane proteins with IgA 71 revealed specificity for a 62-kDa protein. Another of our monoclonal antibodies, IgG 40, precipitated a protein of similar molecular weight. IgA 71 but not IgG 40 also precipitated purified recombinant H. pylori urease. The antigenic specificity of both antibodies was confirmed to be urease by the ability of each to select Escherichia coli clones expressing the H. felis urease genes. The two antibodies were shown to bind nonoverlapping epitopes in a competition enzyme-linked immunosorbent assay. Both IgA 71 and IgG 40 could effectively neutralize H. felis infectivity by incubating the bacteria with the antibodies prior to oral administration to naive mice. The mechanism of protection does not appear to be inhibition of urease activity, as IgA 71 does not inhibit the conversion of urea to ammonia by H. pylori urease in vitro. These results support a protective role for the secretory humoral immune response in Helicobacter immunity and provide further evidence that the urease enzyme can serve as a protective antigen.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Infecções por Helicobacter/prevenção & controle , Helicobacter/imunologia , Urease/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antibacterianos/uso terapêutico , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Sequência de Bases , Primers do DNA/química , Mapeamento de Epitopos , Imunização Passiva , Camundongos , Dados de Sequência Molecular , Urease/antagonistas & inibidores
16.
Cell Immunol ; 191(1): 74-80, 1999 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-9918689

RESUMO

Helicobacter pylori infection of the gastric mucosa can result in gastritis and peptic ulcer disease. Although vaccination can induce protective immunity in animal models of Helicobacter infection, the mechanism(s) of protective immunity has not been fully elucidated. This study was designed to determine whether humoral immune responses are required for protective Helicobacter immunity. IgA-deficient or immunoglobulin-deficient mice were orally immunized against Helicobacter felis and then challenged with live H. felis. Both groups were protected at levels comparable to that of wild-type mice. Additionally, inflammation was equivalent in extent and character between wild-type and antibody-deficient mice. Therefore antibody-independent mechanisms of immunity can protect mice against gastric Helicobacter infection.


Assuntos
Anticorpos Antibacterianos/imunologia , Mucosa Gástrica/imunologia , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Animais , Gastrite/etiologia , Deficiência de IgA/imunologia , Imunidade nas Mucosas , Imunização , Camundongos , Camundongos Endogâmicos C57BL
17.
Gastroenterology ; 110(6): 1770-5, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8964402

RESUMO

BACKGROUND & AIMS: Helicobacter infection of the gastric antrum is responsible for a number of gastric disorders. Antibiotic therapy is lengthy and is not always effective. It has been shown previously that oral immunization against Helicobacter felis in mice can prevent colonization after challenge. The aim of this study was to investigate the efficacy of therapeutic immunization in eradicating an established Helicobacter infection and in reducing gastritis. METHODS: Domestic ferrets, confirmed to be infected with Helicobacter mustelae by gastric endoscopy, were orally immunized with varying doses of purified Helicobacter pylori urease in combination with the mucosal adjuvant cholera toxin. Ferrets were assessed 1 week and 6 weeks after treatment for infection and pathology. RESULTS: Therapeutic immunization eradicated Helicobacter colonization in 30% of all immunized ferrets, although there was no difference in efficacy between the varying doses of antigen tested. The difference was statistically significant when compared with animals administered cholera toxin alone or buffer (P = 0.04). The intensity of inflammation was also significantly reduced in immunized animals (P = 0.0003). CONCLUSIONS: Oral immunization with purified H. pylori urease and cholera toxin can eradicate H. mustelae in a natural host pathogen model. Oral immunization of chronically infected animals markedly reduced gastric inflammation.


Assuntos
Furões , Infecções por Helicobacter/terapia , Infecções por Helicobacter/veterinária , Imunoterapia , Administração Oral , Doenças dos Animais/terapia , Animais , Toxina da Cólera/uso terapêutico , Quimioterapia Combinada , Helicobacter , Infecções por Helicobacter/patologia , Helicobacter pylori/metabolismo , Masculino , Estômago/patologia , Urease/uso terapêutico
18.
J Infect Dis ; 184(3): 308-14, 2001 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-11443556

RESUMO

To determine whether systemic immunization against Helicobacter pylori could be achieved with an adjuvant approved for human use, the efficacy of vaccination with Helicobacter antigen in combination with aluminum hydroxide (AlOH) was evaluated in a murine model of Helicobacter infection. Immunization with antigen and AlOH induced interleukin-5-secreting, antigen-specific T cells, and immunization with antigen and complete Freund's adjuvant induced interferon-gamma-secreting, antigen-specific T cells, as determined by ELISPOT assay. Both immune responses conferred protection after challenge with either H. pylori or H. felis, as confirmed by the complete absence of any bacteria, as assessed by both histology and culture of gastric biopsy samples. Protection was antibody independent, as demonstrated with antibody-deficient muMT mice (immunoglobulin-gene knockout mice), and CD4(+) spleen T cells from immunized mice were sufficient to transfer protective immunity to otherwise immunodeficient rag1(-/-) recipients. These results suggest an alternative and potentially more expeditious strategy for development of a human-use H. pylori vaccine.


Assuntos
Antígenos de Bactérias/imunologia , Linfócitos B/imunologia , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Linfócitos T/imunologia , Transferência Adotiva , Hidróxido de Alumínio , Animais , Antígenos de Bactérias/administração & dosagem , Adjuvante de Freund , Mucosa Gástrica/patologia , Helicobacter/imunologia , Infecções por Helicobacter/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Vacinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA