Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Cell Microbiol ; 10(4): 930-44, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18070119

RESUMO

Pathogenic mycobacteria have the ability to persist in phagocytic cells and to suppress the immune system. The glycolipid lipoarabinomannan (LAM), in particular its mannose cap, has been shown to inhibit phagolysosome fusion and to induce immunosuppressive IL-10 production via interaction with the mannose receptor or DC-SIGN. Hence, the current paradigm is that the mannose cap of LAM is a crucial factor in mycobacterial virulence. However, the above studies were performed with purified LAM, never with live bacteria. Here we evaluate the biological properties of capless mutants of Mycobacterium marinum and M. bovis BCG, made by inactivating homologues of Rv1635c. We show that its gene product is an undecaprenyl phosphomannose-dependent mannosyltransferase. Compared with parent strain, capless M. marinum induced slightly less uptake by and slightly more phagolysosome fusion in infected macrophages but this did not lead to decreased survival of the bacteria in vitro, nor in vivo in zebra fish. Loss of caps in M. bovis BCG resulted in a sometimes decreased binding to human dendritic cells or DC-SIGN-transfected Raji cells, but no differences in IL-10 induction were observed. In mice, capless M. bovis BCG did not survive less well in lung, spleen or liver and induced a similar cytokine profile. Our data contradict the current paradigm and demonstrate that mannose-capped LAM does not dominate the Mycobacterium-host interaction.


Assuntos
Cápsulas Bacterianas/fisiologia , Lipopolissacarídeos/metabolismo , Manose/metabolismo , Mycobacterium/fisiologia , Animais , Cápsulas Bacterianas/metabolismo , Elementos de DNA Transponíveis/genética , Células Dendríticas/metabolismo , Células Dendríticas/microbiologia , Eletroforese em Gel de Poliacrilamida , Feminino , Teste de Complementação Genética , Interações Hospedeiro-Patógeno , Humanos , Immunoblotting , Interleucina-10/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Manose/química , Manose/fisiologia , Manosiltransferases/genética , Manosiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Modelos Moleculares , Mutagênese Insercional , Mutação , Mycobacterium/metabolismo , Infecções por Mycobacterium/metabolismo , Infecções por Mycobacterium/microbiologia , Peixe-Zebra
2.
Aliment Pharmacol Ther ; 21(8): 977-83, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15813833

RESUMO

BACKGROUND: Helicobacter pylori-associated atrophy of the gastric corpus is associated with the presence of anti-canalicular autoantibodies. Also, long-term profound acid suppression in H. pylori-infected subjects may cause atrophic corpus gastritis. AIM: To investigate whether long-term acid suppression by omeprazole leads to antigastric autoantibodies. METHODS: Fifty patients, of which 34 H. pylori-positive on entry of the study, were treated with omeprazole (20-40 mg once daily) for reflux oesophagitis, and were evaluated for anti-gastric autoantibody responses by immunohistochemistry before and after treatment. H. pylori was not eradicated and patients were followed for an average of 6.6 years (range 3-14.1 years). In addition to immunohistochemistry, anti-H(+), K(+)-ATPase reactivity was assessed by Western blot in paired sera of 41 patients (26 H. pylori-positive and 15 uninfected) and results are critically evaluated. RESULTS: In immunohistochemistry, all patients were negative for anti-canalicular autoantibodies when omeprazole therapy started, except for two patients with corpus-predominant gastritis in the presence of H. pylori. One patient, who was H. pylori-negative, newly developed an anti-canalicular antibody response during therapy. CONCLUSIONS: Our results indicate that, as compared with non-infected patients, long-term profound acid suppression therapy in H. pylori-infected gastro-oesophageal reflux disease patients does not increase or accelerate gastric autoimmunity.


Assuntos
Antiulcerosos/uso terapêutico , Autoanticorpos/imunologia , Ácido Gástrico/fisiologia , Refluxo Gastroesofágico/tratamento farmacológico , Refluxo Gastroesofágico/imunologia , Infecções por Helicobacter , Helicobacter pylori , Omeprazol/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Formação de Anticorpos , Western Blotting , Estudos de Coortes , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade
3.
Infect Immun ; 69(12): 7334-40, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11705905

RESUMO

Previously, we have shown that Helicobacter pylori can spontaneously and reversibly change its membrane lipid composition, producing variants with low or high content of lysophospholipids. The "lyso" variant contains a high percentage of lysophospholipids, adheres better to epithelial cells, and releases more proteins such as urease and VacA, compared to the "normal" variant, which has a low content of lysophospholipids. Prolonged growth of the normal variant at pH 3.5, but not under neutral conditions, leads to enrichment of lyso variant colonies, suggesting that the colony switch is relevant to acid adaptation. In this study we show that the change in membrane lipid composition is due to phase variation in the pldA gene. A change in the (C) tract length of this gene results in reversible frameshifts, translation of a full-length or truncated pldA, and the production of active or inactive outer membrane phospholipase A (OMPLA). The role of OMPLA in determining the colony morphology was confirmed by the construction of an OMPLA-negative mutant. Furthermore, variants with an active OMPLA were able to survive acidic conditions better than variants with the inactive form. This explains why the lyso variant is selected at low pH. Our studies demonstrate that phase variation in the pldA gene, resulting in an active form of OMPLA, is important for survival under acidic conditions. We also demonstrated the active OMPLA genotype in fresh isolates of H. pylori from patients referred to gastroscopy for dyspepsia.


Assuntos
Ácidos/farmacologia , Adaptação Biológica/genética , Proteínas da Membrana Bacteriana Externa/genética , Variação Genética , Helicobacter pylori/genética , Fosfolipases A/genética , Mudança da Fase de Leitura do Gene Ribossômico , Helicobacter pylori/enzimologia , Lipopolissacarídeos/química , Lisofosfolipídeos/análise , Fosfolipases A1 , Seleção Genética , Urease/biossíntese
4.
Ned Tijdschr Geneeskd ; 145(36): 1718-22, 2001 Sep 08.
Artigo em Holandês | MEDLINE | ID: mdl-11572169

RESUMO

Adjuvant therapy for severe sepsis and shock can be divided into 4 groups. The first group comprises those compounds with proven efficacy in human studies (activated protein C and recombinant bacterial permeability-increasing protein). The second group includes compounds with potential efficacy (heparin), while the third group represents those with no demonstrated efficacy in randomised clinical trials (tumour necrosis factor and interleukin-1 antibodies and receptor antagonists). The fourth group includes those drugs which have been found to be potentially effective in animal studies, but which have not yet been evaluated in humans (i.e., tyrosine kinase inhibitors, selective inducible nitric oxide synthase inhibitors, polyadenosine-diphosphate-ribose-polymerase and caspase III (apoptosis) inhibitors). Formal clinical comparisons between the various treatment options are necessary to assist the clinician in selecting the appropriate form of therapy.


Assuntos
Cuidados Críticos/métodos , Sepse/tratamento farmacológico , Choque Séptico/tratamento farmacológico , Animais , Anti-Inflamatórios/uso terapêutico , Anticoagulantes/uso terapêutico , Proteínas Sanguíneas/uso terapêutico , Quimioterapia Combinada , Inibidores Enzimáticos/uso terapêutico , Heparina/uso terapêutico , Humanos , Hidrocortisona/uso terapêutico , Proteínas de Membrana/uso terapêutico , Proteína C/uso terapêutico , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptores de Interleucina/antagonistas & inibidores , Sepse/mortalidade , Índice de Gravidade de Doença , Choque Séptico/mortalidade , Fator de Necrose Tumoral alfa/uso terapêutico
5.
Biochem Cell Biol ; 79(4): 449-59, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11527214

RESUMO

Helicobacter pylori is a widespread Gram-negative bacterium responsible for the onset of various gastric pathologies and cancers in humans. A familiar trait of H. pylori is the production of cell-surface lipopolysaccharides (LPSs; O-chain --> core --> lipid A) with O-chain structures analogous to some mammalian histo-blood-group antigens, those being the Lewis determinants (Lea, Leb, Lex, sialyl Lex, Ley) and blood groups A and linear B. Some of these LPS antigens have been implicated as autoimmune, adhesion, and colonization components of H. pylori pathogenic mechanisms. This article describes the chemical structures of LPSs from H. pylori isolated from subjects with no overt signs of disease. Experimental data from chemical- and spectroscopic-based studies unanimously showed that these H. pylori manufactured extended heptoglycans composed of 2- and 3-linked D-glycero-alpha-D-manno-heptopyranose units and did not express any blood-group O-antigen chains. The fact that another H. pylori isolate with a similar LPS structure was shown to be capable of colonizing mice indicates that H. pylori histo-blood-group structures are not an absolute prerequisite for colonization in the murine model also. The absence of O-chains with histo-blood groups may cause H. pylori to become inept in exciting an immune response. Additionally, the presence of elongated heptoglycans may impede exposure of disease-causing outer-membrane antigens. These factors may render such H. pylori incapable of creating exogenous contacts essential for pathogenesis of severe gastroduodenal diseases and suggest that histo-blood groups in the LPS may indeed play a role in inducing a more severe H. pylori pathology.


Assuntos
Helicobacter pylori/metabolismo , Antígenos do Grupo Sanguíneo de Lewis , Polissacarídeos/metabolismo , Sequência de Carboidratos , Eletroforese em Gel de Poliacrilamida , Helicobacter pylori/patogenicidade , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Polissacarídeos/química
6.
Infect Immun ; 69(4): 2708-13, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11254638

RESUMO

Infection with Helicobacter pylori has been associated with induction of autoantibodies that cross-react with the gastric mucosa. There have been discordant reports as to whether or not these autoantibodies arise due to molecular mimicry between H. pylori and host cell antigens on parietal cells. In this study, we investigated whether molecular mimicry by H. mustelae causes autoantibodies in infected ferrets. Serum from H. mustelae-infected ferrets reacted with parietal cells in the ferret gastric mucosa but not with duodenal or colonic mucosa. These sera did not react with the blood group A epitope on erythrocytes or H. mustelae lipopolysaccharide, and absorption with H. mustelae whole cells or red blood cells did not remove autoantibodies. In conclusion, ferrets naturally infected with H. mustelae generate antibodies that react with parietal cells, but these autoantibodies are not due to molecular mimicry.


Assuntos
Autoanticorpos/sangue , Mucosa Gástrica/imunologia , Infecções por Helicobacter/imunologia , Sistema ABO de Grupos Sanguíneos/imunologia , Animais , Anticorpos Antibacterianos/sangue , Furões , Gastrite Atrófica/etiologia , ATPase Trocadora de Hidrogênio-Potássio/imunologia , Antígenos CD15/imunologia , Lipopolissacarídeos/imunologia , Coelhos
7.
FEMS Microbiol Lett ; 196(2): 235-8, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11267785

RESUMO

Helicobacter pylori infection results in chronic gastritis, which is initiated by the release of cytokines like interleukin (IL)-12 and IL-8 from mononuclear cells, and IL-8 from gastric epithelial cells. The severity of gastritis is influenced both by host factors and by bacterial factors such as the Cag proteins and the vacuolating cytotoxin VacA. Amounts of IL-12 and IL-8 produced by monocytic THP-1 cells differed considerably between the eight H. pylori isolates tested, but in contrast to H. pylori-induced IL-8 production by gastric epithelial cells, did not correlate to the Cag and VacA types of the strains. Apparently, in addition to Cag and VacA, other bacterial factors determine the extent in which H. pylori induced IL production in monocytes.


Assuntos
Helicobacter pylori/imunologia , Interleucina-12/biossíntese , Interleucina-8/biossíntese , Monócitos/imunologia , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/fisiologia , Células Cultivadas , Células Epiteliais/metabolismo , Mucosa Gástrica/citologia , Mucosa Gástrica/imunologia , Antígenos HLA-D , Helicobacter pylori/genética , Humanos , Imunidade nas Mucosas , Interleucina-12/análise , Interleucina-8/análise , Virulência/genética
8.
Gastroenterology ; 120(2): 377-86, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11159878

RESUMO

BACKGROUND & AIMS: The proton pump H(+),K(+)-adenosine triphosphatase (H(+),K(+)-ATPase) of parietal cells is the major humoral autoantigen in both human and experimental autoimmune gastritis (AIG) characterized by an inflammatory infiltrate in the gastric mucosa and loss of parietal cells. The aim of this study was to detect H(+),K(+)-ATPase-specific T cells in the gastric mucosa of patients with AIG and to define their functional properties. METHODS: In vivo-activated T cells from the infiltrates of the gastric mucosa of 5 patients with AIG were isolated and cloned. The ability of gastric T-cell clones to proliferate and to produce cytokines in response to H(+),K(+)-ATPase, as well as their expression of B-cell help, perforin-mediated cytotoxicity, and Fas-Fas ligand-mediated apoptosis in target cells, were assessed. RESULTS: A proportion (25%) of the CD4(+) clones from the gastric corpus of AIG patients proliferated in response to porcine H(+),K(+)-ATPase. Most of these clones (88%) showed a Th1 profile, whereas a few secreted both Th1 and Th2 cytokines. Virtually all of the H(+),K(+)-ATPase-specific clones produced tumor necrosis factor alpha and provided substantial help for B-cell immunoglobulin production, and most of them expressed perforin-mediated cytotoxicity against antigen-presenting cells and induced Fas-Fas ligand-mediated apoptosis in target cells. CONCLUSIONS: Activation of proton pump-specific Th1 cytotoxic/proapoptotic T cells in the gastric mucosa can represent an effector mechanism for the target cell destruction in AIG.


Assuntos
Doenças Autoimunes/imunologia , Gastrite/imunologia , ATPase Trocadora de Hidrogênio-Potássio/imunologia , Linfócitos T Citotóxicos/imunologia , Células Th1/imunologia , Adulto , Autoantígenos/imunologia , Doenças Autoimunes/patologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Biópsia , Morte Celular/imunologia , Células Clonais , Epitopos , Proteína Ligante Fas , Feminino , Mucosa Gástrica/imunologia , Gastrite/patologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Humanos , Imunoglobulina G/biossíntese , Glicoproteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Perforina , Proteínas Citotóxicas Formadoras de Poros , Células Th1/enzimologia
9.
Trends Microbiol ; 8(12): 565-70, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11115753

RESUMO

In mimicry with human gastric epithelial cells, the lipopolysaccharide of Helicobacter pylori expresses Lewis blood group antigens. Recent data suggest that molecular mimicry does not promote immune evasion, nor does it lead to induction of autoantibodies, but that H. pylori Lewis X mediates adhesion to gastric epithelial cells and is essential for colonization.


Assuntos
Infecções por Helicobacter/microbiologia , Helicobacter pylori/patogenicidade , Antígenos do Grupo Sanguíneo de Lewis/fisiologia , Antígenos O/fisiologia , Aderência Bacteriana/fisiologia , Sequência de Carboidratos , Helicobacter pylori/imunologia , Helicobacter pylori/fisiologia , Humanos , Antígenos do Grupo Sanguíneo de Lewis/química , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Mimetismo Molecular , Dados de Sequência Molecular , Antígenos O/química , Antígenos O/imunologia , Virulência
10.
Infect Immun ; 68(10): 5928-32, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10992504

RESUMO

Helicobacter pylori NCTC 11637 lipopolysaccharide (LPS) expresses the human blood group antigens Lewis x (Le(x)), Le(y), and H type I. In this report, we demonstrate that the H type I epitope displays high-frequency phase variation. One variant expressed Le(x) and Le(y) and no H type I as determined by serology; this switch was reversible. Insertional mutagenesis in NCTC 11637 of JHP563 (a poly(C) tract containing an open reading frame homologous to glycosyltransferases) yielded a transformant with a serotype similar to the phase variant. Structural analysis of the NCTC 11637 LPS confirmed the loss of the H type I epitope. Sequencing of JHP563 in strains NCTC 11637, an H type I-negative variant, and an H type I-positive switchback variant showed a C14 (gene on), C13 (gene off), and C14 tract, respectively. Inactivation of strain G27, which expresses Le(x), Le(y), H type I, and Le(a), yielded a transformant that expressed Le(x) and Le(y). We conclude that JHP563 encodes a beta3-galactosyltransferase involved in the biosynthesis of H type I and Le(a) and that phase variation in H type I is due to C-tract changes in this gene. A second H type I-negative variant (variant 3a) expressed Le(x) and Le(a) and had lost both H type I and Le(y) expression. Inactivation of HP093-HP094 resulted in a transformant expressing Le(x) and lacking Le(y) and H type I. Structural analysis of a mutant LPS confirmed the serological data. We conclude that the HP093-HP094 alpha2-fucosyltransferase (alpha2-FucT) gene product is involved in the biosynthesis of both Le(y) and Le(x). Finally, we inactivated HP0379 in strain 3a. The transformant had lost both Le(x) and Le(a) expression, which demonstrates that the HP0379 gene product is both an alpha3- and an alpha4-FucT. Our data provide understanding at the molecular level of how H. pylori is able to diversify in the host, a requirement likely essential for successful colonization and transmission.


Assuntos
Epitopos , Helicobacter pylori/imunologia , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Lipopolissacarídeos/classificação , Lipopolissacarídeos/imunologia , Anticorpos Monoclonais/imunologia , Sequência de Carboidratos , Galactosiltransferases/química , Galactosiltransferases/genética , Galactosiltransferases/metabolismo , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Humanos , Antígenos do Grupo Sanguíneo de Lewis/química , Lipopolissacarídeos/química , Dados de Sequência Molecular , Mutagênese Insercional , Reação em Cadeia da Polimerase , Análise de Sequência de DNA
12.
Eur J Biochem ; 267(2): 305-20, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10632700

RESUMO

This study describes the molecular makeup of the cell-wall lipopolysaccharides (LPSs) (O-chain polysaccharide-->core oligosaccharide-->lipid A) from five Helicobacter pylori strains: H. pylori 26695 and J99, the complete genome sequences of which have been published, the established mouse model Sydney strain (SS1), and the symptomatic strains P466 and UA915. All chemical and serological experiments were performed on the intact LPSs. H. pylori 26695 and SS1 possessed either a low-Mr semi-rough-form LPS carrying mostly a single Ley type-2 blood-group determinant in the O-chain region covalently attached to the core oligosaccharide or a high-Mr smooth-form LPS, as did strain J99, with an elongated partially fucosylated type-2 N-acetyllactosamine (polyLacNAc) O-chain polymer, terminated mainly by a Lex blood-group determinant, connected to the core oligosaccharide. In the midst of semi-rough-form LPS glycoforms, H. pylori 26695 and SS1 also expressed in the O-chain region a difucosylated antigen, alpha-L-Fucp(1-3)-alpha-L-Fucp(1-4)-beta-D-GlcpNAc, and the cancer-cell-related type-1 or type-2 linear B-blood-group antigen, alpha-D-Galp(1-3)-beta-D-Galp(1-3 or 4)-beta-D-GlcpNAc. The LPS of H. pylori strain P466 carried the cancer-associated type-2 sialyl Lex blood-group antigen, and the LPS from strain UA915 expressed a type-1 Leb blood-group unit. These findings should aid investigations that focus on identifying and characterizing genes responsible for LPS biosynthesis in genomic strains 26695 and J99, and in understanding the role of H. pylori LPS in animal model studies. The LPSs from the H. pylori strains studied to date were grouped into specific glycotype families.


Assuntos
Helicobacter pylori/química , Antígenos CD15/química , Lipopolissacarídeos/química , Lipopolissacarídeos/classificação , Animais , Sequência de Carboidratos , Helicobacter pylori/classificação , Helicobacter pylori/genética , Helicobacter pylori/imunologia , Antígenos do Grupo Sanguíneo de Lewis/química , Lipopolissacarídeos/análise , Espectroscopia de Ressonância Magnética , Camundongos/microbiologia , Dados de Sequência Molecular , Oligossacarídeos/química , Especificidade da Espécie
14.
Infect Immun ; 67(10): 5361-6, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10496917

RESUMO

The lipopolysaccharide (LPS) of Helicobacter pylori expresses the Lewis x (Lex) and/or Ley antigen. We have shown previously that H. pylori LPS displays phase variation whereby an Lex-positive strain yields variants with different LPS serotypes, for example, Lex plus Ley or nonfucosylated polylactosamine. H. pylori has two alpha3-fucosyltransferase genes that both contain poly(C) tracts. We now demonstrate that these tracts can shorten or lengthen randomly, which results in reversible frameshifting and inactivation of the gene products. We provide genetic and serological evidence that this mechanism causes H. pylori LPS phase variation and demonstrate that the on or off status of alpha3-fucosyltransferase genes determines the LPS serotypes of phase variants and clinical isolates. The role of the alpha3-fucosyltransferase gene products in determining the LPS serotype was confirmed by structural-chemical analysis of alpha3-fucosyltransferase knockout mutants. The data also show that the two alpha3-fucosyltransferase genes code for enzymes with different fine specificities, and we propose the names futA and futB to designate the orthologs of the H. pylori 26695 alpha3-fucosyltransferase genes HP0379 and HP0651, respectively. The data also show that the alpha3-fucosylation precedes alpha2-fucosylation [corrected], an order of events opposite to that which prevails in mammals. Finally, the data provide an understanding at the molecular level of the mechanisms underlying LPS diversity in H. pylori, which may play an important role in adaptation to the host.


Assuntos
Fucosiltransferases/genética , Helicobacter pylori/patogenicidade , Lipopolissacarídeos/química , Poli C/química , Fucosiltransferases/fisiologia , Antígenos do Grupo Sanguíneo de Lewis/análise , Antígenos CD15/análise , Mutação
16.
Antimicrob Agents Chemother ; 42(12): 3073-8, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9835493

RESUMO

Antibiotics with different mechanisms of action may vary with respect to their effects on the release and immunostimulatory activities of cell wall fragments from gram-positive bacteria. Therefore, after Staphylococcus aureus was cultured for 4 h in the absence of antibiotics (control) and in the presence of beta-lactam antibiotics (imipenem, flucloxacillin, or cefamandole) and protein synthesis-inhibiting antibiotics (erythromycin, clindamycin, or gentamicin), the lipoteichoic acid (LTA) and peptidoglycan (PG) levels in the bacterial supernatants were measured. beta-Lactam antibiotics greatly enhanced the release of LTA and PG (4- to 9-fold and 60- to 85-fold, respectively), whereas protein synthesis inhibitors did not affect PG release and even inhibited the release of LTA compared to the amount of LTA released in control cultures. The capacity of beta-lactam supernatants to stimulate the production of tumor necrosis factor alpha and interleukin-10 in human whole blood was significantly higher than that of protein synthesis inhibitor or control supernatants; the amounts of these cytokines released were directly proportional to the concentrations of PG and LTA in the supernatants. Enzymatic degradation of PG in the supernatants indicated that PG was mainly responsible for the observed biological reactivity.


Assuntos
Antibacterianos/farmacologia , Lipopolissacarídeos/metabolismo , Peptidoglicano/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Ácidos Teicoicos/metabolismo , Parede Celular/imunologia , Parede Celular/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Técnicas In Vitro , Interleucina-10/sangue , Lactamas , Lipopolissacarídeos/imunologia , Testes de Sensibilidade Microbiana , Peptidoglicano/imunologia , Inibidores da Síntese de Proteínas/farmacologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/metabolismo , Ácidos Teicoicos/imunologia , Fator de Necrose Tumoral alfa/biossíntese
17.
Gastroenterology ; 115(2): 340-7, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9679039

RESUMO

BACKGROUND & AIMS: A subgroup of Helicobacter pylori-infected patients develops autoantibodies to gastric parietal cell canaliculi. The aim of this study was to define the unknown autoantigen. METHODS: We screened 72 H. pylori-infected patients, 5 patients with autoimmune gastritis, and 36 healthy controls for immunoglobulin G autoantibodies to canaliculi by immunohistochemistry. The antigen specificity was determined by immunoprecipitation of the murine gastric H+,K+-adenosine triphosphatase (H+,K+-ATPase) expressed in oocytes and by immunoblotting on human gastric membranes from the body mucosa. RESULTS: Autoantibodies specific for the conformational peptides of the H+,K+-ATPase were detected in 3% (1/36) of controls, in all patients with autoimmune gastritis (5/5), in 25% (18/72) of H. pylori-infected patients, and in 47% (15/32) of the infected patients with anticanalicular autoantibodies. No other major autoantigen was identified. Atrophy in the gastric body mucosa was found in 60% (9/15) of infected patients with both anticanalicular and anti-H+,K+-ATPase antibodies, but only in 13% (5/37) of infected patients lacking both autoantibodies (P < 0.01). CONCLUSIONS: The gastric H+,K+-ATPase is a major autoantigen in H. pylori-associated antigastric autoimmunity. Thus, anti-H+,K+-ATPase autoantibodies, which are closely linked to classical autoimmune gastritis, are also significant indicators for body mucosa atrophy in chronic H. pylori gastritis.


Assuntos
Autoantígenos/imunologia , Gastrite/imunologia , Gastrite/microbiologia , ATPase Trocadora de Hidrogênio-Potássio/imunologia , Infecções por Helicobacter/imunologia , Helicobacter pylori , Estômago/enzimologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Atrofia , Autoanticorpos/análise , Doença Crônica , Feminino , Mucosa Gástrica/patologia , Gastrite/patologia , Humanos , Isoantígenos/análise , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Masculino , Pessoa de Meia-Idade , Células Parietais Gástricas/imunologia
19.
J Biol Chem ; 273(19): 11533-43, 1998 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-9565568

RESUMO

Previous structural investigations performed on the lipopolysaccharides (LPSs) from the human gastric pathogen Helicobacter pylori have revealed that these cell surface glycan molecules express type 2 partially fucosylated, glucosylated, or galactosylated N-acetyllactosamine O antigen chains (O-chains) of various lengths, which may or may not be terminated at the nonreducing end by Lewis X (Lex) and/or Ley blood group epitopes in mimicry of human cell surface glycoconjugates and glycolipids. Subsequently, serological experiments with commercially available Lewis-specific monoclonal antibodies also have recognized the presence of Lex and Ley blood group antigens in H. pylori but, in addition, have indicated the presence of type 1 chain Lea, Leb, and Led (H-type 1) blood group epitopes in some H. pylori strains. To confirm their presence, structural studies and additional serological experiments were undertaken on H. pylori strains suspected of carrying type 1 chain epitopes. These investigations revealed that the O-chain region of H. pylori strain UA948 carried both Lea (type 1) and Lex (type 2) blood group determinants. The O-chain from H. pylori UA955 LPS expressed the terminal Lewis disaccharide (type 1 chain) and Lex and Ley antigens (type 2). The O-chain of H. pylori J223 LPS carried the type 1 chain precursor Lec, the H-1 epitope (Led, type 1 chain) and an elongated nonfucosylated type 2 N-acetyllactosamine chain (i antigen). Thus, O-chains from H. pylori LPSs can also express fucosylated type 1 sequences, and the LPS from a single H. pylori strain may carry O-chains with type 1 and 2 Lewis blood groups simultaneously. That monoclonal antibodies putatively specific for the Leb determinant can detect glycan substructures (Le disaccharide, Lec, and Led) of Leb indicates their nonspecificity. The expression of both type 1 and 2 Lewis antigens by H. pylori LPSs mimics the cell surface glycomolecules present in both the gastric superficial (which expresses mainly type 1 determinants) and the superficial and glandular epithelium regions (both of which express predominantly type 2 determinants). Therefore, each H. pylori strain may have a different niche within the gastric mucosa, and each individual LPS blood group antigen may have a dissimilar role in H. pylori adaptation.


Assuntos
Mucosa Gástrica/imunologia , Helicobacter pylori/imunologia , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Lipopolissacarídeos/imunologia , Mimetismo Molecular , Sequência de Carboidratos , Cromatografia Gasosa , Células Epiteliais/imunologia , Helicobacter pylori/química , Humanos , Lipopolissacarídeos/química , Espectrometria de Massas , Dados de Sequência Molecular
20.
Infect Immun ; 66(2): 444-7, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9453593

RESUMO

The in vitro binding of surface-exposed material and outer membrane proteins of Helicobacter pylori to high-molecular-weight salivary mucin was studied. We identified a 16-kDa surface protein which adhered to high-molecular-weight salivary mucin. This protein binds specifically to sulfated oligosaccharide structures such as sulfo-Lewis a, sulfogalactose and sulfo-N-acetyl-glucosamine on mucin. Sequence analysis of the protein proved that it was identical to the N-terminal amino acid sequence of neutrophil-activating protein. Moreover, this adhesin was able to bind to Lewis x blood group antigen.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/fisiologia , Helicobacter pylori/fisiologia , Mucinas/metabolismo , Saliva/microbiologia , Sequência de Aminoácidos , Antígenos CD18/fisiologia , Humanos , Antígenos do Grupo Sanguíneo de Lewis/metabolismo , Dados de Sequência Molecular , Peso Molecular , Oligossacarídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA