Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 3804, 2023 06 26.
Article in English | MEDLINE | ID: mdl-37365163

ABSTRACT

The interleukin-1 family members, IL-1ß and IL-18, are processed into their biologically active forms by multi-protein complexes, known as inflammasomes. Although the inflammasome pathways that mediate IL-1ß processing in myeloid cells have been defined, those involved in IL-18 processing, particularly in non-myeloid cells, are still not well understood. Here we report that the host defence molecule NOD1 regulates IL-18 processing in mouse epithelial cells in response to the mucosal pathogen, Helicobacter pylori. Specifically, NOD1 in epithelial cells mediates IL-18 processing and maturation via interactions with caspase-1, instead of the canonical inflammasome pathway involving RIPK2, NF-κB, NLRP3 and ASC. NOD1 activation and IL-18 then help maintain epithelial homoeostasis to mediate protection against pre-neoplastic changes induced by gastric H. pylori infection in vivo. Our findings thus demonstrate a function for NOD1 in epithelial cell production of bioactive IL-18 and protection against H. pylori-induced pathology.


Subject(s)
Epithelial Cells , Helicobacter Infections , Interleukin-18 , Nod1 Signaling Adaptor Protein , Animals , Mice , Epithelial Cells/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori , Inflammasomes/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction , Nod1 Signaling Adaptor Protein/metabolism
2.
Mol Immunol ; 42(8): 879-85, 2005 May.
Article in English | MEDLINE | ID: mdl-15829277

ABSTRACT

Toll-like receptor (TLR) molecules play a frontline role in the defence of the host against infection by microbial pathogens. These molecules, together with the recently described Nod family proteins, have been shown to trigger innate immune responses in host cells via the recognition of highly conserved microbial structures. TLR4, which is the best-characterised of these "pathogen-recognition molecules" (PRMs), was the first to be shown to recognise a specific microbial component: the lipopolysaccharide (LPS) from Gram-negative bacteria. The molecular specificities of the remaining PRMs have, in nearly all cases, now also been elucidated. Host cells belonging to the myeloid cell lineage are known to be particularly responsive to these microbial constituents. Conversely, other cell types such as epithelial cells, were generally thought to be hypo-responsive to stimulation by such molecules. New evidence suggests that these cells are in fact likely to play a fundamental role in host defence against pathogenic micro-organisms. Indeed, epithelial cells afford an initial barrier against the host microflora, and appear to be able to differentiate between pathogenic and commensal micro-organisms. This review article will discuss current knowledge regarding innate immune responses in epithelial and myeloid cells to the model non-invasive pathogen, Helicobacter pylori, which is a major cause of upper gastrointestinal tract disease in humans.


Subject(s)
Helicobacter Infections/immunology , Helicobacter pylori/immunology , Immunity, Innate/immunology , Animals , Dendritic Cells/immunology , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Humans , Macrophages/immunology , Monocytes/immunology
3.
Infect Immun ; 70(8): 4581-90, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12117971

ABSTRACT

Helicobacter pylori infection causes active chronic inflammation with a continuous recruitment of neutrophils to the inflamed gastric mucosa. To evaluate the role of endothelial cells in this process, we have examined adhesion molecule expression and chemokine and cytokine production from human umbilical vein endothelial cells stimulated with well-characterized H. pylori strains as well as purified proteins. Our results indicate that endothelial cells actively contribute to neutrophil recruitment, since stimulation with H. pylori bacteria induced upregulation of the adhesion molecules VCAM-1, ICAM-1, and E-selectin as well as the chemokines interleukin 8 (IL-8) and growth-related oncogene alpha (GRO-alpha) and the cytokine IL-6. However, there were large variations in the ability of the different H. pylori strains to stimulate endothelial cells. These interstrain variations were seen irrespective of whether the strains had been isolated from patients with duodenal ulcer disease or asymptomatic carriers and were not solely related to the expression of known virulence factors, such as the cytotoxin-associated gene pathogenicity island, vacuolating toxin A, and Lewis blood group antigens. In addition, one or several unidentified proteins which act via NF-kappaB activation seem to induce endothelial cell activation. In conclusion, human endothelial cells produce neutrophil-recruiting factors and show increased adhesion molecule expression after stimulation with certain H. pylori strains. These effects probably contribute to the continuous recruitment of neutrophils to H. pylori-infected gastric mucosa and may also contribute to tissue damage and ulcer formation.


Subject(s)
Endothelium, Vascular/immunology , Helicobacter pylori/immunology , Intercellular Signaling Peptides and Proteins , Adult , Bacterial Adhesion , Bacterial Proteins/immunology , Cells, Cultured , Chemokine CCL5/biosynthesis , Chemokine CXCL1 , Chemokine CXCL10 , Chemokines, CXC/biosynthesis , Chemotactic Factors/biosynthesis , E-Selectin/biosynthesis , Endothelium, Vascular/cytology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Growth Substances/biosynthesis , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Interleukin-8/genetics , NF-kappa B/metabolism , Vascular Cell Adhesion Molecule-1/biosynthesis
4.
Infect Immun ; 69(8): 5186-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11447204

ABSTRACT

The effect of low-dose antigen exposure on the development of immunity to Helicobacter pylori infection was studied in outbred mice. Animals that were primed with a subinfectious number of H. pylori bacteria exhibited significantly lower bacterial loads after challenge with an infectious dose of pathogen (versus controls, P < 0.05).


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Animals , Antibodies, Bacterial/blood , Helicobacter Infections/blood , Helicobacter Infections/pathology , Interferon-gamma/analysis , Interleukin-4/analysis , Mice , Ribosomal Proteins/immunology , Stomach/microbiology , Stomach/pathology
6.
Antimicrob Agents Chemother ; 44(10): 2623-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10991835

ABSTRACT

The main objectives of this study were to determine whether the nitroreductase enzyme encoded by the rdxA gene of Helicobacter pylori was responsible for reductive activation of nitrofurantoin and whether a triple-therapy regimen with nitrofurantoin was able to eradicate metronidazole-sensitive and -resistant H. pylori infections from mice. The susceptibilities to nitrofurantoin of parent and isogenic rdxA mutant strains (three pairs), as well as a series of matched metronidazole-sensitive and -resistant strains isolated from mice (30) and patients (20), were assessed by agar dilution determination of the MIC. Groups of mice colonized with the metronidazole-sensitive H. pylori SS1 strain or a metronidazole-resistant rdxA SS1 mutant were treated with either metronidazole or nitrofurantoin as part of a triple-therapy regimen. One month after the completion of treatment the mice were sacrificed and their stomachs were cultured for H. pylori. The nitrofurantoin MICs for all strains tested were between 0.5 and 4.0 microg/ml. There was no significant difference between the susceptibility to nitrofurantoin of the parental strains and those of respective rdxA mutants or between those of matched metronidazole-sensitive and -resistant H. pylori isolates. The regimen with metronidazole eradicated infection from all eight SS1-infected mice and from one of eight mice inoculated with the rdxA mutant (P < or =0.001). The regimen with nitrofurantoin failed to eradicate infection from any of the six SS1-infected mice (P < or =0.001) and cleared infection from one of seven mice inoculated with the rdxA mutant. These results demonstrate that, despite the good in vitro activity of nitrofurantoin against H. pylori and the lack of cross-resistance between metronidazole and nitrofurantoin, eradication regimens involving nitrofurantoin are unable to eradicate either metronidazole-sensitive or -resistant H. pylori infections from mice.


Subject(s)
Anti-Infective Agents, Urinary/therapeutic use , Antitrichomonal Agents/pharmacology , Helicobacter Infections/drug therapy , Helicobacter pylori/drug effects , Metronidazole/pharmacology , Nitrofurantoin/therapeutic use , Animals , Bacterial Proteins/genetics , Cloning, Molecular , DNA Primers , Drug Resistance, Microbial , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Membrane Proteins/genetics , Mice , Microbial Sensitivity Tests , Mutation/genetics , Oxidation-Reduction , Reverse Transcriptase Polymerase Chain Reaction
7.
J Pathol ; 191(3): 333-40, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10878557

ABSTRACT

Experimental infection of mice with Helicobacter felis reproduces many aspects of the gastritis observed in Helicobacter pylori-infected humans. The development of gastric inflammatory lesions in chronically infected inbred mice is host-dependent; in BALB/c mice, gastric B-cell MALT lymphomas were observed, whilst other murine hosts (e.g. C57BL/6) developed severe glandular hyperplasia. The aims of this investigation were to characterize and immunophenotype Helicobacter-induced inflammatory lesions in mice with an outbred genetic background. Swiss mice (n=10 per group) were either inoculated with a suspension of H. felis or left untreated. H. felis-inoculated mice and age-matched control animals were killed 13 months later. The severity of gastric inflammatory lesions in the animals was graded and the number and distribution of B (CD45R(+)) and T (CD3(+)) lymphocytes in lymphoid tissues was determined by immunohistochemistry. Compared with control mice, animals with long-term H. felis infection developed severe hyperplastic gastritis (0.80+/-0.63 vs. 2.7+/-0.68), with epithelial dedifferentiation (0. 40+/-0.52 vs. 2.3+/-0.82) and lengthening of the pits and glands (0. 46+/-0.05 vs. 0.8+/-0.19). Gastric CD45R(+) and CD3(+) lymphocyte scores were significantly elevated (r=0.803) in infected animals, while lymphoepithelial lesions and polymorphonuclear leucocyte infiltrates were absent. Although prominent lymphoid follicles were present in the tissues of all infected animals, and in one control animal, only a proportion (55%) of the mucosal follicles had a dominant B-cell phenotype (defined as > or =75% CD45R(+) labelling), and all were poorly labelled with anti-mouse immunoglobulin antibodies. It was concluded that the lesions in outbred Swiss mice differed from B-cell MALT lymphomas. In contrast to inbred mice, outbred animals developed both glandular and lymphoid tissue lesions to chronic H. felis infection. It is suggested that the default T-helper phenotype of the host influences glandular lesion formation or B-cell lymphomagenesis in this model of infection.


Subject(s)
Gastritis/microbiology , Helicobacter Infections/pathology , Lymphoid Tissue/pathology , Stomach/pathology , Animals , B-Lymphocyte Subsets/immunology , CD3 Complex/analysis , Chronic Disease , Gastritis/immunology , Gastritis/pathology , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Hyperplasia/immunology , Hyperplasia/microbiology , Hyperplasia/pathology , Immunoenzyme Techniques , Immunoglobulin A/biosynthesis , Immunoglobulin G/biosynthesis , Leukocyte Common Antigens/analysis , Lymphoid Tissue/immunology , Lymphoid Tissue/microbiology , Lymphoma, B-Cell, Marginal Zone/microbiology , Mice , Species Specificity , Specific Pathogen-Free Organisms , Stomach/immunology , Stomach/microbiology , T-Lymphocytes, Helper-Inducer/immunology
8.
J Feline Med Surg ; 2(1): 19-27, 2000 Mar.
Article in English | MEDLINE | ID: mdl-11716588

ABSTRACT

The types of helicobacter which are found in the stomachs of carnivorous pets, especially cats, have been traditionally referred to as 'gastric helicobacter-like organisms' (GHLOs). These are microaerophilic, Gram-negative, spiral bacteria with multiple terminal flagellae and are endowed with high-level urease activity which allows them to survive in an acidic environment. Certain species have one or more periplasmic fibrils. The two GHLOs most commonly found in cats are Helicobacter felis and a species related to H heilmannii which was recently cultured from dogs. All phenotypic and genotypic (16S RNA gene sequences) evidence suggests that both of these bacteria belong in the genus Helicobacter. Whether or not helicobacters can be transmitted to humans from carnivorous pets is controversial but the recent discovery of H pylori -infected cats may be evidence of an animal reservoir for this pathogen. Although the role of H pylori in inducing antral gastritis and perpetuating pyloric ulcers in humans is well established, whether or not Helicobacter spp are causally involved in any feline gastric inflammatory conditions is unknown.


Subject(s)
Cat Diseases/microbiology , Helicobacter Infections/veterinary , Helicobacter/isolation & purification , Stomach/microbiology , Animals , Cat Diseases/diagnosis , Cat Diseases/epidemiology , Cats , Gastritis/microbiology , Gastritis/pathology , Gastritis/veterinary , Helicobacter/pathogenicity , Helicobacter Infections/diagnosis , Helicobacter Infections/epidemiology , Helicobacter Infections/microbiology , Helicobacter pylori , Prevalence
9.
J Bacteriol ; 181(23): 7314-22, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10572136

ABSTRACT

Arginase of the Helicobacter pylori urea cycle hydrolyzes L-arginine to L-ornithine and urea. H. pylori urease hydrolyzes urea to carbon dioxide and ammonium, which neutralizes acid. Both enzymes are involved in H. pylori nitrogen metabolism. The roles of arginase in the physiology of H. pylori were investigated in vitro and in vivo, since arginase in H. pylori is metabolically upstream of urease and urease is known to be required for colonization of animal models by the bacterium. The H. pylori gene hp1399, which is orthologous to the Bacillus subtilis rocF gene encoding arginase, was cloned, and isogenic allelic exchange mutants of three H. pylori strains were made by using two different constructs: 236-2 and rocF::aphA3. In contrast to wild-type (WT) strains, all rocF mutants were devoid of arginase activity and had diminished serine dehydratase activity, an enzyme activity which generates ammonium. Compared with WT strain 26695 of H. pylori, the rocF::aphA3 mutant was approximately 1, 000-fold more sensitive to acid exposure. The acid sensitivity of the rocF::aphA3 mutant was not reversed by the addition of L-arginine, in contrast to the WT, and yielded a approximately 10, 000-fold difference in viability. Urease activity was similar in both strains and both survived acid exposure equally well when exogenous urea was added, indicating that rocF is not required for urease activity in vitro. Finally, H. pylori mouse-adapted strain SS1 and the 236-2 rocF isogenic mutant colonized mice equally well: 8 of 9 versus 9 of 11 mice, respectively. However, the rocF::aphA3 mutant of strain SS1 had moderately reduced colonization (4 of 10 mice). The geometric mean levels of H. pylori recovered from these mice (in log(10) CFU) were 6.1, 5.5, and 4.1, respectively. Thus, H. pylori rocF is required for arginase activity and is crucial for acid protection in vitro but is not essential for in vivo colonization of mice or for urease activity.


Subject(s)
Arginase/metabolism , Bacterial Proteins , Helicobacter pylori/enzymology , Mice/microbiology , Agmatine/metabolism , Alleles , Animals , Arginase/genetics , Arginine/metabolism , Blotting, Southern , Cloning, Molecular , Deamination , Gene Silencing , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Mutagenesis , Urea/pharmacology , Urease/metabolism
10.
J Antimicrob Chemother ; 43(6): 753-8, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10404313

ABSTRACT

It was recently demonstrated that inactivation of the rdxA gene, which encodes an oxygen-insensitive NADPH nitroreductase, is associated with the development of resistance to metronidazole by Helicobacter pylori. In order to further evaluate the contribution of rdxA to metronidazole resistance, the sequence of the rdxA gene was determined for a series of metronidazole-sensitive and -resistant isolates derived from a single, metronidazole-sensitive strain using an H. pylori mouse model. These strains were cultured from the stomachs of mice experimentally infected with H. pylori strain SS1 and then treated orally with metronidazole. The sequence of the rdxA gene of all 10 sequenced metronidazole-sensitive and two (7%) of the 27 metronidazole-resistant isolates was identical to that of the parental strain. In contrast, the rdxA gene of the other 25 metronidazole-resistant isolates contained between one and three frameshift or missense mutations. This suggests that while the development of metronidazole resistance in H. pylori is frequently associated with mutational inactivation of the rdxA gene, other mechanisms of resistance are likely to exist in this bacterium.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Helicobacter pylori/drug effects , Membrane Proteins/genetics , Metronidazole/pharmacology , Bacterial Proteins/physiology , Drug Resistance, Microbial/genetics , Evolution, Molecular , Helicobacter pylori/genetics , Humans , Membrane Proteins/physiology , Mutation, Missense
11.
Mol Microbiol ; 33(2): 350-62, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10411751

ABSTRACT

Helicobacter felis has been used extensively in animal model studies of gastric Helicobacter infections. Attempts to manipulate H. felis genetically have, however, been unsuccessful and, consequently, little is known about the pathogenic mechanisms of this bacterium. In common with other Helicobacter spp., H. felis is a highly motile organism. To characterize the flagellar structures responsible for this motility, we cloned and sequenced the two flagellin-encoding genes, flaA and flaB, from H. felis. These genes encode two flagellin proteins that are expressed simultaneously under the control of putative sigma28 and sigma54 promoters respectively. Isogenic mutants of H. felis in flaA and flaB were generated by electroporation-mediated allelic disruption and replacement, showing for the first time that H. felis could be manipulated genetically. Both types of H. felis flagellin mutants exhibited truncated flagella and were poorly motile. H. felis flaA mutants were unable to colonize the gastric mucosa in a mouse infection model.


Subject(s)
Alleles , Flagellin/genetics , Helicobacter/genetics , Animals , Blotting, Western , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Glycosylation , Mice , Microscopy, Electron , Molecular Weight , Mutagenesis , Phylogeny , Promoter Regions, Genetic , Protein Processing, Post-Translational
12.
Mol Microbiol ; 31(5): 1359-72, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10200957

ABSTRACT

Constitutive expression of gamma-glutamyltranspeptidase (GGT) activity is common to all Helicobacter pylori strains, and is used as a marker for identifying H. pylori isolates. Helicobacter pylori GGT was purified from sonicated extracts of H. pylori strain 85P by anion exchange chromatography. The N-terminal amino acid sequences of two of the generated endo-proteolysed peptides were determined, allowing the cloning and sequencing of the corresponding gene from a genomic H. pylori library. The H. pylori ggt gene consists of a 1681 basepair (bp) open reading frame encoding a protein with a signal sequence and a calculated molecular mass of 61 kDa. Escherichia coli clones harbouring the H. pylori ggt gene exhibited GGT activity at 37 degrees C, in contrast to E. coli host cells (MC1061, HB101), which were GGT negative at 37 degrees C. GGT activity was found to be constitutively expressed by similar genes in Helicobacter felis, Helicobacter canis, Helicobacter bilis, Helicobacter hepaticus and Helicobacter mustelae. Western immunoblots using rabbit antibodies raised against a His-tagged-GGT recombinant protein demonstrated that H. pylori GGT is synthesized in both H. pylori and E. coli as a pro-GGT that is processed into a large and a small subunit. Deletion of a 700 bp fragment within the GGT-encoding gene of a mouse-adapted H. pylori strain (SS1) resulted in mutants that were GGT negative yet grew normally in vitro. These mutants, however, were unable to colonize the gastric mucosa of mice when orally administered alone or together (co-infection) with the parental strain. These results demonstrate that H. pylori GGT activity has an essential role for the establishment of the infection in the mouse model, demonstrating for the first time a physiological role for a bacterial GGT enzyme.


Subject(s)
Gastric Mucosa/microbiology , Helicobacter pylori/enzymology , Helicobacter pylori/pathogenicity , gamma-Glutamyltransferase/physiology , Amino Acid Sequence , Animals , Blotting, Western , Cloning, Molecular , Escherichia coli/genetics , Genetic Vectors , Mice , Models, Genetic , Molecular Sequence Data , Mutagenesis , Plasmids , Sequence Homology, Amino Acid , Time Factors , gamma-Glutamyltransferase/isolation & purification
13.
Antimicrob Agents Chemother ; 43(4): 777-81, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10103180

ABSTRACT

The Helicobacter pylori SS1 mouse model was used to characterize the development of resistance in H. pylori after treatment with metronidazole monotherapy and to examine the effect of prior exposure to metronidazole on the efficacy of a metronidazole-containing eradication regimen. Mice colonized with the metronidazole-sensitive H. pylori SS1 strain were treated for 7 days with either peptone trypsin broth or the mouse equivalent of 400 mg of metronidazole once a day or three times per day (TID). In a separate experiment, H. pylori-infected mice were administered either peptone trypsin broth or the mouse equivalent of 400 mg of metronidazole TID for 7 days, followed 1 month later by either peptone trypsin broth or the mouse equivalent of 20 mg of omeprazole, 250 mg of clarithromycin, and 400 mg of metronidazole twice a day for 7 days. At least 1 month after the completion of treatment, the mice were sacrificed and their stomachs were cultured for H. pylori. The susceptibilities of isolates to metronidazole were assessed by agar dilution determination of the MICs. Mixed populations of metronidazole-resistant and -sensitive strains were isolated from 70% of mice treated with 400 mg of metronidazole TID. The ratio of resistant to sensitive strains was 1:100, and the MICs for the resistant strains varied from 8 to 64 micrograms/ml. In the second experiment, H. pylori was eradicated from 70% of mice treated with eradication therapy alone, compared to 25% of mice pretreated with metronidazole (P < 0.01). Mice still infected after treatment with metronidazole and eradication therapy contained mixed populations of metronidazole-resistant and -sensitive isolates in a ratio of 1:25. These results demonstrate that H. pylori readily acquires resistance to metronidazole in vivo and that prior exposure of the organism to metronidazole is associated with failure of eradication therapy. H. pylori-infected mice provide a suitable model for the study of resistance mechanisms in H. pylori and will be useful in determining optimal regimens for the eradication of resistant strains.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Helicobacter Infections/drug therapy , Metronidazole/therapeutic use , Animals , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Drug Resistance, Microbial/physiology , Helicobacter Infections/microbiology , Helicobacter Infections/transmission , Helicobacter pylori/drug effects , Metronidazole/pharmacology , Mice , Treatment Outcome
14.
Biochim Biophys Acta ; 1388(2): 465-77, 1998 Nov 10.
Article in English | MEDLINE | ID: mdl-9858781

ABSTRACT

The properties of Helicobacter pylori arginase activity in metabolically competent cells and lysates were investigated with the aim of obtaining a better understanding of the nitrogen metabolism of the bacterium. One-dimensional 1H- and 13C-nuclear magnetic resonance spectroscopy, spectrophotometry, radio tracer analysis and protein purification techniques were employed to characterize in situ the first step in the utilization of l-arginine by the bacterium. Arginase activity was associated with the cell-envelope fraction obtained by centrifugation of lysates. A Km of 22+/-3 mM was determined for the enzyme activity, and differences of Vmax were observed between strains. Divalent cations stimulated arginase activity, and the most potent activators were Co2+>Ni2+>Mn2+. The activity was highly specific for l-arginine and did not catabolize analogs recognized by other arginases of prokaryote and eukaryote origin. The Ki of several inhibitors was measured and served also to characterize the enzyme activity. The presence of bicarbonate enhanced the hydrolysis of l-arginine in cell suspensions, but not in lysates or semi-purified enzyme preparations. Amino acid sequence analyses revealed important differences between the deduced structures of H. pylori arginase and those of other organisms. This finding was consistent with experimental data which showed that H. pylori arginase has unique properties.


Subject(s)
Arginase/chemistry , Helicobacter pylori/enzymology , Arginine/metabolism , Bacterial Proteins/chemistry , Biological Transport , Cations, Divalent/pharmacology , Enzyme Inhibitors/pharmacology , Isoelectric Point , Kinetics , Magnetic Resonance Spectroscopy , Molecular Weight , Ornithine/pharmacology , Sequence Homology, Amino Acid , Substrate Specificity
15.
Infect Immun ; 66(4): 1349-55, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9529052

ABSTRACT

A model permitting the establishment of persistent Helicobacter pylori infection in mice was recently described. To evaluate murine immune responses to H. pylori infection, specific-pathogen-free Swiss mice (n = 50) were intragastrically inoculated with 1.2 x 10(7) CFU of a mouse-adapted H. pylori isolate (strain SS1). Control animals (n = 10) received sterile broth medium alone. Animals were sacrificed at various times, from 3 days to 16 weeks postinoculation (p.i.). Quantitative culture of gastric tissue samples from inoculated mice demonstrated bacterial loads of 4.0 x 10(4) to 8 x 10(6) CFU per g of tissue in the animals. Infected mice had H. pylori-specific immunoglobulin M (IgM) and IgG antibodies in serum (at day 3 p.i.) and IgG and IgA antibodies in their gastric contents (weeks 4 and 16 p.i.) and saliva (week 16 p.i.). Mucosal IgM antibodies were not detected. Histological examination of the gastric mucosae from control and infected mice revealed mild chronic gastritis, characterized by the presence of polymorphoneutrophil cell infiltrates and submucosal lymphoid aggregates, in infected animals at 16 weeks p.i. Differences in the quantities of IgG1 and IgG2a subclass antibodies detected in the sera of mouse strains (Swiss, BALB/c, and C57BL/6) infected by H. pylori suggested that host factors influence the immune responses induced against this bacterium in the host. In conclusion, immune responses to H. pylori infection in mice, like those in chronically infected humans, appear to be ineffective in resolving the infection.


Subject(s)
Helicobacter Infections/immunology , Helicobacter pylori , Animals , Antibodies, Bacterial/biosynthesis , Chronic Disease , Helicobacter Infections/pathology , Immunoglobulin A/biosynthesis , Immunoglobulin G/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Specific Pathogen-Free Organisms
16.
Gastroenterology ; 113(1): 185-94, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9207277

ABSTRACT

BACKGROUND & AIMS: Orogastric immunization of mice with Helicobacter antigens, together with a mucosal adjuvant (cholera toxin), has been shown to confer immunity in the Helicobacter felis infection model. The aim of the study was to investigate the humoral immune responses associated with immunity and to compare these with responses in H. felis-infected mice. METHODS: Enzyme-linked immunoassays were used to characterize the antibody-secreting cells and antibodies present at mucosal and systemic sites in mice. Animals were immunized orally with either whole-cell H. felis sonicates or Helicobacter pylon urease or heat-shock proteins. RESULTS: Infection of mice with H. felis preferentially induced the recruitment of plasma cells committed to immunoglobulin (Ig) A synthesis in salivary gland and gastric tissues. Antigen-specific IgA was the major antibody class detected in mucosal secretions recovered from these tissues. In contrast, immunization of mice against H. felis infection induced the proliferation of large numbers of IgG-secreting cells, as well as the synthesis of local IgG antibodies, in the gastric mucosa of the animals. Protection against H. felis infection occurred in the absence of gastric IgA responses in sonicate-immunized mice. CONCLUSIONS: It is proposed that locally synthesized specific IgG antibodies contribute to immunity against gastric Helicobacter infection.


Subject(s)
Gastric Mucosa/immunology , Gastritis/immunology , Gastritis/microbiology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Helicobacter/immunology , Immunoglobulin G/immunology , Animals , Antibody-Producing Cells/immunology , Helicobacter Infections/microbiology , Immunization , Immunoglobulin A/biosynthesis , Immunoglobulin A/immunology , Immunoglobulin G/biosynthesis , Mice
17.
Vet Res ; 28(3): 207-15, 1997.
Article in English | MEDLINE | ID: mdl-9208441

ABSTRACT

As a result of phylogenic studies using new molecular biology techniques and fundamental experimental studies, we now know more about helicobacteria in domestic carnivores, their morphologic characteristics, their taxonomia and more important we know more about their ecological niche. Few clinical studies have been carried out, but the ones that have been undertaken are interesting in that they confirm the extensive prevalence of Helicobacter infections in domestic carnivores and underline their role in the genesis of the inflammatory gastropathies observed in these species. Finally, recent observations have demonstrated the ubiquitous character of these helicobacteria by showing their presence in the stomach of man, dog and cat. This ubiquitous character has led some scientists to consider the potential zoonotic risk of the human infection by Helicobacter heilmannii, felis or pylori.


Subject(s)
Helicobacter Infections/veterinary , Helicobacter/pathogenicity , Stomach Diseases/veterinary , Animals , Animals, Domestic , Cat Diseases , Cats , Dog Diseases , Dogs , Helicobacter/ultrastructure , Helicobacter Infections/pathology , Helicobacter Infections/transmission , Helicobacter pylori , Humans , Stomach Diseases/microbiology , Stomach Diseases/pathology , Zoonoses
19.
Proc Natl Acad Sci U S A ; 92(14): 6499-503, 1995 Jul 03.
Article in English | MEDLINE | ID: mdl-7604021

ABSTRACT

Helicobacter pylori is an important etiologic agent of gastroduodenal disease. In common with other organisms, H. pylori bacteria express heat shock proteins that share homologies with the GroES-GroEL class of proteins from Escherichia coli. We have assessed the heat shock proteins of H. pylori as potential protective antigens in a murine model of gastric Helicobacter infection. Orogastric immunization of mice with recombinant H. pylori GroES- and GroEL-like proteins protected 80% (n = 20) and 70% (n = 10) of animals, respectively, from a challenge dose of 10(4) Helicobacter felis bacteria (compared to control mice, P = 0.0042 and P = 0.0904, respectively). All mice (n = 19) that were immunized with a dual antigen preparation, consisting of H. pylori GroES-like protein and the B subunit of H. pylori urease, were protected against infection. This represented a level of protection equivalent to that provided by a sonicated Helicobacter extract (P = 0.955). Antibodies directed against the recombinant H. pylori antigens were predominantly of the IgG1 class, suggesting that a type 2 T-helper cell response was involved in protection. This work reports a protein belonging to the GroES class of heat shock proteins that was shown to induce protective immunity. In conclusion, GroES-like and urease B-subunit proteins have been identified as potential components of a future H. pylori subunit vaccine.


Subject(s)
Chaperonin 10/immunology , Gastric Mucosa/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Helicobacter pylori/pathogenicity , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/blood , Enzyme-Linked Immunosorbent Assay , Escherichia coli/metabolism , Gastric Mucosa/pathology , Helicobacter Infections/prevention & control , Helicobacter pylori/metabolism , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Mice , Recombinant Proteins/immunology
20.
Mol Microbiol ; 14(5): 959-74, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7715457

ABSTRACT

All Helicobacter pylori isolates synthesize a 54 kDa immunodominant protein that was reported to be associated with the nickel-dependent urease of H. pylori. This protein was recently recognized as a homologue of the heat-shock protein of the GroEL class. The gene encoding the GroEL-like protein of H. pylori (HspB) was cloned (pILL689) and was shown to belong to a bicistronic operon including the hspA and hspB genes. In Escherichia coli, the constitutive expression of the hspA and hspB genes was initiated from a promoter located within an IS5 insertion element that mapped upstream to the two open reading frames (ORFs). IS5 was absent from the H. pylori genome, and was thus acquired during the cosmid cloning process. hspA and hspB encoded polypeptides of 118 and 545 amino acid residues, corresponding to calculated molecular masses of 13.0 and 58.2 kDa, respectively. Amino acid sequence comparison studies revealed that, although H. pylori HspA and HspB proteins were highly similar to their bacterial homologues, the H. pylori HspA featured a striking motif at the C-terminus. This unique motif consists of a series of cysteine and histidine residues resembling a nickel-binding domain, which is not present in any of the other bacterial GroES homologues so far characterized. When the pILL689 recombinant plasmid was introduced together with the H. pylori urease gene cluster (pILL763) into an E. coli host strain, an increase of urease activity was observed. This suggested a close interaction between the HspA and HspB proteins and the urease enzyme, and a possible role for HspA in the chelation of nickel ions. The genes encoding each of the HspA and HspB polypeptides were cloned, expressed independently as proteins fused to the maltose-binding protein (MBP) and purified in large scale. The MBP-HspA and MBP-HspB fusion proteins were shown to retain their antigenic properties. Both HspA and HspB represent antigens that are specifically recognized by the sera from H. pylori-infected patients. Whereas HspB was known to be immunogenic in humans, this is the first demonstration that HspA per se is also immunogenic.


Subject(s)
Genes, Bacterial , Heat-Shock Proteins/genetics , Helicobacter pylori/genetics , Amino Acid Sequence , Base Sequence , Chaperonin 60/genetics , Chaperonin 60/immunology , Chaperonin 60/metabolism , Cosmids , DNA Primers/genetics , DNA, Bacterial/genetics , Gene Expression , Heat-Shock Proteins/immunology , Heat-Shock Proteins/metabolism , Helicobacter pylori/immunology , Helicobacter pylori/metabolism , Humans , Molecular Sequence Data , Multigene Family , Nickel/metabolism , Plasmids/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Restriction Mapping , Sequence Homology, Amino Acid , Transcription, Genetic , Urease/genetics , Urease/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...