Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Exp Med ; 197(7): 813-22, 2003 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-12668646

RESUMO

Helicobacter pylori causes one of the most common, chronic bacterial infections and is a primary cause of severe gastric disorders. To unravel the bacterial factors necessary for the process of gastric colonization and pathogenesis, signature tagged mutagenesis (STM) was adapted to H. pylori. The Mongolian gerbil (Meriones unguiculatus) was used as model system to screen a set of 960 STM mutants. This resulted in 47 H. pylori genes, assigned to 9 different functional categories, representing a set of biological functions absolutely essential for gastric colonization, as verified and quantified for many mutants by competition experiments. Identification of previously known colonization factors, such as the urease and motility functions validated this method, but also novel and several hypothetical genes were found. Interestingly, a secreted collagenase, encoded by hp0169, could be identified and functionally verified as a new essential virulence factor for H. pylori stomach colonization. Furthermore, comB4, encoding a putative ATPase being part of a DNA transformation-associated type IV transport system of H. pylori was found to be absolutely essential for colonization, but natural transformation competence was apparently not the essential function. Thus, this first systematic STM application identified a set of previously unknown H. pylori colonization factors and may help to potentiate the development of novel therapies against gastric Helicobacter infections.


Assuntos
Genes Bacterianos , Helicobacter pylori/genética , Estômago/microbiologia , Animais , Colagenases/genética , DNA Bacteriano/química , Flagelina/genética , Biblioteca Gênica , Gerbillinae , Helicobacter pylori/fisiologia , Humanos , Mutação , Fases de Leitura Aberta , Transformação Bacteriana , Urease/genética
2.
Proteomics ; 9(6): 1582-92, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19253298

RESUMO

Because gastric infection by Helicobacter pylori takes place via the oral route, possible interactions of this bacterium with human salivary proteins could occur. By using modified 1- and 2-D bacterial overlay, binding of H. pylori adhesins BabA and SabA to the whole range of salivary proteins was explored. Bound salivary receptor molecules were identified by MALDI-MS and by comparison to previously established proteome maps of whole and glandular salivas. By use of adhesin-deficient mutants, binding of H. pylori to MUC7 and gp-340 could be linked to the SabA and BabA adhesins, respectively, whereas binding to MUC5B was associated with both adhesins. Binding of H. pylori to the proline-rich glycoprotein was newly detected and assigned to BabA adhesin whereas the SabA adhesin was found to mediate binding to newly detected receptor molecules, including carbonic anhydrase VI, secretory component, heavy chain of secretory IgA1, parotid secretory protein and zinc-alpha(2)-glycoprotein. Some of these salivary glycoproteins are known to act as scavenger molecules or are involved in innate immunity whereas others might come to modify the pathogenetic properties of this organism. In general, this 2-D bacterial overlay technique represents a useful supplement in adhesion studies of bacteria with complex protein mixtures.


Assuntos
Adesinas Bacterianas/metabolismo , Eletroforese em Gel Bidimensional/métodos , Glicoproteínas/análise , Helicobacter pylori/metabolismo , Proteoma/análise , Receptores de Superfície Celular/análise , Saliva/metabolismo , Aderência Bacteriana , Western Blotting , Fluoresceína-5-Isotiocianato , Fluorescência , Glicoproteínas/isolamento & purificação , Humanos , Lectinas/metabolismo , Peso Molecular , Ligação Proteica , Proteínas e Peptídeos Salivares/isolamento & purificação
3.
Infect Immun ; 77(9): 3782-90, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19546190

RESUMO

The gram-negative gastric pathogen Helicobacter pylori is equipped with an extraordinarily large set of outer membrane proteins (OMPs), whose role in the infection process is not well understood. The Hop (Helicobacter outer membrane porins) and Hor (Hop-related proteins) groups constitute a large paralogous family consisting of 33 members. The OMPs AlpA, AlpB, BabA, SabA, and HopZ have been identified as adhesins or adherence-associated proteins. To better understand the relevance of these and other OMPs during infection, we analyzed the expression of eight different omp genes (alpA, alpB, babA, babB, babC, sabA, hopM, and oipA) in a set of 200 patient isolates, mostly from symptomatic children or young adults. Virtually all clinical isolates produced the AlpA and AlpB proteins, supporting their essential function. All other OMPs were produced at extremely variable rates, ranging from 35% to 73%, indicating a function in close adaptation to the individual host or gastric niche. In 11% of the isolates, BabA was produced, and SabA was produced in 5% of the isolates, but the strains failed to bind their cognate substrates. Interleukin-8 (IL-8) expression in gastric cells was strictly dependent on the presence of the cag pathogenicity island, whereas the presence of OipA clearly enhanced IL-8 production. The presence of the translocated effector protein CagA correlated well with BabA and OipA production. In conclusion, we found unexpectedly diverse omp expression profiles in individual H. pylori strains and hypothesize that this reflects the selective pressure for adhesion, which may differ across different hosts as well as within an individual over time.


Assuntos
Proteínas da Membrana Bacteriana Externa/análise , Helicobacter pylori/química , Adesinas Bacterianas/análise , Antígenos de Bactérias/análise , Aderência Bacteriana , Proteínas de Bactérias/análise , Linhagem Celular Tumoral , Colágeno Tipo IV/metabolismo , Helicobacter pylori/fisiologia , Humanos , Interleucina-8/biossíntese , Estômago/microbiologia
4.
PLoS Pathog ; 2(10): e110, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17121461

RESUMO

Adherence of Helicobacter pylori to inflamed gastric mucosa is dependent on the sialic acid-binding adhesin (SabA) and cognate sialylated/fucosylated glycans on the host cell surface. By in situ hybridization, H. pylori bacteria were observed in close association with erythrocytes in capillaries and post-capillary venules of the lamina propria of gastric mucosa in both infected humans and Rhesus monkeys. In vivo adherence of H. pylori to erythrocytes may require molecular mechanisms similar to the sialic acid-dependent in vitro agglutination of erythrocytes (i.e., sialic acid-dependent hemagglutination). In this context, the SabA adhesin was identified as the sialic acid-dependent hemagglutinin based on sialidase-sensitive hemagglutination, binding assays with sialylated glycoconjugates, and analysis of a series of isogenic sabA deletion mutants. The topographic presentation of binding sites for SabA on the erythrocyte membrane was mapped to gangliosides with extended core chains. However, receptor mapping revealed that the NeuAcalpha2-3Gal-disaccharide constitutes the minimal sialylated binding epitope required for SabA binding. Furthermore, clinical isolates demonstrated polymorphism in sialyl binding and complementation analysis of sabA mutants demonstrated that polymorphism in sialyl binding is an inherent property of the SabA protein itself. Gastric inflammation is associated with periodic changes in the composition of mucosal sialylation patterns. We suggest that dynamic adaptation in sialyl-binding properties during persistent infection specializes H. pylori both for individual variation in mucosal glycosylation and tropism for local areas of inflamed and/or dysplastic tissue.


Assuntos
Adesinas Bacterianas/metabolismo , Helicobacter pylori/fisiologia , Hemaglutininas/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Polissacarídeos/metabolismo , Adesinas Bacterianas/genética , Adsorção , Animais , Antígenos de Bactérias/metabolismo , Aderência Bacteriana , Sítios de Ligação , Ligação Competitiva , Capilares , Eritrócitos/metabolismo , Eritrócitos/microbiologia , Gangliosídeos/metabolismo , Mucosa Gástrica/irrigação sanguínea , Mucosa Gástrica/microbiologia , Deleção de Genes , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/metabolismo , Hemaglutinação , Humanos , Técnicas In Vitro , Macaca mulatta , Oligossacarídeos/metabolismo , Antígeno Sialil Lewis X , Vênulas
5.
Clin Gastroenterol Hepatol ; 5(1): 49-58, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17157077

RESUMO

BACKGROUND & AIMS: The blood grou. METHODS: We compared the ability of published PCR-based methods to assess BabA status with BabA immunoblotting and Lewis b (Le(b)) binding activity assays. We also used immunoblotting to examine the relationship between clinical presentation and levels of BabA expression. RESULTS: Immunoblotting and Le(b) binding assays for 80 strains revealed 3 levels of BabA expression: BabA high producers (BabA-H) with Le(b) binding activity, BabA low producers (BabA-L) without Le(b) binding activity, and BabA-negative. BabA-negative strains lacked the babA gene. PCR methods to determine BabA status yielded poor results. babA1 sequences were never detected. BabA expression was examined in 250 strains from Western countries and 270 strains from East Asia. The results failed to confirm any relationship between triple-positive status (cagA-positive/vacA s1/BabA-H) and clinical outcome. BabA-negative strains typically were cagA-negative/vacA s2 and were associated with gastritis. BabA-L strains showed a higher level of mucosal injury and were associated more frequently with duodenal ulcer and gastric cancer than the other groups. CONCLUSIONS: Information gained from currently used PCR-based methods must be interpreted with caution. Le(b) binding activity does not accurately reflect the severity of mucosal damage or the clinical outcome. Quantitation of BabA expression revealed that Le(b)-nonbinding BabA-L strains are associated with higher levels of mucosal injury and clinical outcome.


Assuntos
Adesinas Bacterianas/genética , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Helicobacter pylori/genética , Adenocarcinoma/genética , Adesinas Bacterianas/análise , Adesinas Bacterianas/metabolismo , Primers do DNA , Úlcera Duodenal/genética , Gastrite/genética , Helicobacter pylori/isolamento & purificação , Helicobacter pylori/metabolismo , Humanos , Immunoblotting , Antígenos do Grupo Sanguíneo de Lewis/imunologia , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo , Índice de Gravidade de Doença , Neoplasias Gástricas/genética
6.
J Biol Chem ; 282(9): 6242-54, 2007 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-17202133

RESUMO

Following adhesion of Helicobacter pylori to gastric epithelial cells, intracellular signaling leads to cytokine production, which causes H. pylori-related gastric injury. Two adjacent homologous genes (alpA and alpB), which encode H. pylori outer membrane proteins, are thought to be associated with adhesion and cytokine induction. We co-cultured gastric epithelial cells with wild type H. pylori strains and their corresponding alpA/alpB-deleted mutants (DeltaalpAB). Results were confirmed by complementation. Flow cytometry confirmed that AlpAB was involved in cellular adhesion. Deletion of alpAB reduced interleukin (IL)-6 induction in gastric epithelial cells. Deletion of alpAB reduced IL-8 induction with East Asian but not with Western strains. All AlpAB-positive strains tested activated the extracellular signal-regulated kinase, c-Fos, and cAMP-responsive element-binding protein. Activation of the Jun-N-terminal kinase, c-Jun, and NF-kappaB was exclusive to AlpAB from East Asian strains. DeltaalpAB mutants poorly colonized the stomachs of C57BL/6 mice and were associated with lower mucosal levels of KC and IL-6. Our results suggest that AlpAB may induce gastric injury by mediating adherence to gastric epithelial cells and by modulating proinflammatory intracellular signaling cascades. Known geographical differences in H. pylori-related clinical outcomes may relate to differential effects of East Asian and Western types of AlpAB on NF-kappaB-related proinflammatory signaling pathways.


Assuntos
Adesinas Bacterianas/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Células Epiteliais/microbiologia , Helicobacter pylori/química , Transdução de Sinais , Adesinas Bacterianas/genética , Animais , Proteínas da Membrana Bacteriana Externa/genética , Linhagem Celular Tumoral , Citocinas , Geografia , Humanos , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutação , Estômago/microbiologia
7.
J Bacteriol ; 188(3): 882-93, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16428391

RESUMO

Helicobacter pylori is one of the most diverse bacterial species known. A rational basis for this genetic variation may be provided by its natural competence for genetic transformation and high-frequency recombination. Many bacterial competence systems have homology with proteins that are involved in the assembly of type IV pili and type II secretion systems. In H. pylori, DNA uptake relies on a transport system related to type IV secretion systems (T4SS) designated the comB system. The prototype of a T4SS in Agrobacterium tumefaciens consists of 11 VirB proteins and VirD4, which form the core unit necessary for the delivery of single proteins or large nucleoprotein complexes into target cells. In the past we identified proteins ComB4 and ComB7 through ComB10 as being involved in the process of DNA uptake in H. pylori. In this study we identified and functionally characterized further (T4SS-homologous) components of the comB transformation competence system. By combining computer prediction modeling, experimental topology determination, generation of knockout strains, and genetic complementation studies we identified ComB2, ComB3, and ComB6 as essential components of the transformation apparatus, structurally and functionally homologous to VirB2, VirB3, and VirB6, respectively. comB2, comB3, and comB4 are organized as a separate operon. Thus, for the H. pylori comB system, all T4SS core components have been identified except for homologues to VirB1, VirD4, VirB5, and VirB11.


Assuntos
Proteínas de Bactérias/fisiologia , Helicobacter pylori/fisiologia , Transformação Bacteriana/genética , Fatores de Virulência , Proteínas de Bactérias/química , Biologia Computacional , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Helicobacter pylori/genética , Fases de Leitura Aberta/genética
8.
Helicobacter ; 11(3): 196-207, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16684268

RESUMO

BACKGROUND: The colonization of the gastric mucosa with Helicobacter pylori is accompanied by elevated levels of proinflammatory cytokines, such as interleukin-1 (IL-1), IL-6, and IL-8. The aim of our study was to determine the mechanisms of IL-6 stimulation in phagocytes upon H. pylori infection. MATERIALS AND METHODS: We investigated the secretion of IL-6 by different professional phagocytes from murine and human origin, including granulocyte- and monocyte-like cells and macrophages derived from human peripheral blood monocytes (PBMCs). The influence of viability, phagocytosis, and the impact of different subcellular fractions of H. pylori bacteria were evaluated. RESULTS: IL-6 levels induced by H. pylori were low in cell lines derived from murine and human monocytes and in human granulocyte-like cells. By contrast, macrophages derived from human PBMCs were highly responsive to both H. pylori and Escherichia coli. IL-6 induction was blocked by inhibition of actin-dependent processes prior to infection with H. pylori, but not with E. coli or E. coli lipopolysaccharide (LPS). Using cell fractionation, the most activity was found in the H. pylori membrane. H. pylori LPS exhibited a 10(3)- to 10(4)-fold lower biologic activity than E. coli LPS, suggesting a minor role for toll-like receptor 4 (TLR4)-mediated signalling from the exterior. CONCLUSIONS: From these data, we conclude that macrophages may be a major source of IL-6 in the gastric mucosa upon H. pylori infection. The IL-6 induction by H. pylori in these cells is a multifactorial process, which requires the uptake and presumably degradation of H. pylori bacteria.


Assuntos
Mucosa Gástrica/imunologia , Helicobacter pylori/imunologia , Interleucina-6/metabolismo , Macrófagos/metabolismo , Fagocitose , Animais , Linhagem Celular , Células HL-60 , Helicobacter pylori/patogenicidade , Humanos , Ativação de Macrófagos , Macrófagos/imunologia , Camundongos
9.
Am J Gastroenterol ; 101(1): 36-44, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16405531

RESUMO

OBJECTIVES: We tested whether the interaction between host gastric Le(x) antigen and the SabA protein of H. pylori determined gastric colonization density. METHODS: A total of 145 H. pylori-infected patients were assessed for their bacterial density and gastric Le(b) and sialyl-Le(x) expression. Their corresponding H. pylori isolates were tested for babA2 and sabA genotype by PCR. The sabA-genopositive PCR products were sequenced to check for mutations affecting SabA expression. The BabA and SabA expressions of each isolate were confirmed by Western blotting. RESULTS: All 145 H. pylori isolates were babA2-genopositive and expressed BabA. There were 116 (80%) sabA-genopositive isolates, but only 45 (31%) of the isolates expressed SabA. Sequence of sabA-genopositive PCR products was achieved in 92 isolates, of which 60% had regular CT repeat-pairs and the other 40% had a unique deletion of the CT repeats. Neither the deletion nor the different CT repeat-pairs in the sabA region were totally correlated with SabA expression, defined by Western blotting. H. pylori density was higher in those expressing gastric sialyl-Le(x) antigen (which interacts with SabA) (p < 0.001) only in those patients expressing weak or no gastric Le(b) antigen (which would interact with BabA), not in those with evident expression of gastric Le(b) antigen. CONCLUSIONS: In Taiwan, H. pylori isolates are 100% BabA-positive, but only 31% of them express SabA. The interaction between gastric sialyl-Le(x) and SabA of H. pylori determines the colonization density of patients expressing gastric Le(b) weakly or not at all.


Assuntos
Adesinas Bacterianas/genética , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Antígenos do Grupo Sanguíneo de Lewis/metabolismo , Antígenos CD15/metabolismo , Estômago/microbiologia , Adulto , Idoso , Análise de Variância , Antígenos de Bactérias/análise , Aderência Bacteriana/genética , Sequência de Bases , Estudos de Coortes , Feminino , Regulação Bacteriana da Expressão Gênica , Helicobacter pylori/isolamento & purificação , Helicobacter pylori/patogenicidade , Humanos , Concentração de Íons de Hidrogênio , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Probabilidade , Sensibilidade e Especificidade
10.
Infect Immun ; 74(7): 3845-52, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16790756

RESUMO

Maintaining metal homeostasis is crucial for the adaptation of Helicobacter pylori to the gastric environment. Iron, copper, and nickel homeostasis has recently been demonstrated to be required for the establishment of H. pylori infection in animal models. Here we demonstrate that the HP0969-0971 gene cluster encoding the Czc-type metal export pump homologs HP0969, HP0970, and the H. pylori-specific protein HP0971 forms part of a novel H. pylori metal resistance determinant, which is required for gastric colonization and for the modulation of urease activity. Insertional mutagenesis of the HP0971, HP0970, or HP0969 genes in H. pylori reference strain 26695 resulted in increased sensitivity to cadmium, zinc, and nickel (czn), suggesting that the encoded proteins constitute a metal-specific export pump. Accordingly, the genes were designated cznC (HP0971), cznB (HP0970), and cznA (HP0969). The CznC and CznA proteins play a predominant role in nickel homeostasis, since only the cznC and cznA mutants but not the cznB mutant displayed an 8- to 10-fold increase in urease activity. Nickel-specific affinity chromatography demonstrated that recombinant versions of CznC and CznB can bind to nickel and that the purified CznB protein interacted with cadmium and zinc, since both metals competitively inhibited nickel binding. Finally, single cznA, cznB, and cznC mutants did not colonize the stomach in a Mongolian gerbil-based animal model. This demonstrates that the metal export functions of H. pylori cznABC are essential for gastric colonization and underlines the extraordinary importance of metal ion homeostasis for the survival of H. pylori in the gastric environment.


Assuntos
Proteínas de Bactérias/fisiologia , Cádmio/metabolismo , Proteínas de Transporte de Cátions/fisiologia , Mucosa Gástrica/microbiologia , Helicobacter pylori/metabolismo , Níquel/metabolismo , Urease/metabolismo , Zinco/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Transporte de Cátions/genética , Gerbillinae , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/enzimologia , Helicobacter pylori/crescimento & desenvolvimento , Helicobacter pylori/patogenicidade , Mutação
11.
Int J Med Microbiol ; 295(5): 317-24, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16173498

RESUMO

The adherence of the human gastric pathogen Helicobacter pylori to the gastric mucosa is widely assumed to play a substantial role in initial colonization and long-term persistence in the human stomach. In the past, a couple of putative adhesins were identified, most of which were members of the large outer membrane protein (OMP) family of H. pylori. Among these, the BabA protein was shown to recognize the Leb antigen, which is presented as a dominant surface structure in the gastric mucosa. The SabA adhesin binds to sialylated antigens, which are up-regulated in inflamed gastric tissue. Other OMPs, such as AlpAB or HopZ were also shown to be involved in binding to gastric epithelial cells, but the corresponding receptors are unknown. The aim of this review is to summarize recent data giving new insights in binding specificities of H. pylori adhesins and their role in pathogenesis and adaptation to the host.


Assuntos
Aderência Bacteriana/fisiologia , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/patogenicidade , Adaptação Fisiológica , Adesinas Bacterianas/fisiologia , Animais , Proteínas da Membrana Bacteriana Externa/fisiologia , Helicobacter pylori/fisiologia , Helicobacter pylori/ultraestrutura , Humanos
12.
Glycobiology ; 15(7): 700-8, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15716466

RESUMO

The microaerophilic bacterium Helicobacter pylori is well established for its role in development of different gastric diseases. Bacterial adhesins and corresponding binding sites on the epithelial surface allow H. pylori to colonize the gastric tissue. In this investigation, the adhesion of H. pylori to dot blot arrays of natural glycoproteins and neoglycoproteins was studied. Adhesion was detected by overlay with fluorescence-labeled bacteria on immobilized (neo)glycoproteins. The results confirmed the interaction between the adhesin BabA and the H-1-, Lewis b-, and related fucose-containing antigens. In addition, H. pylori bound to terminal alpha2-3-linked sialic acids as previously described. The use of a sabA mutant and sialidase treatment of glycoconjugate arrays showed that the adherence of H. pylori to laminin is mediated by the sialic acid-binding adhesin, SabA. The adhesion to salivary mucin MUC5B is mainly associated with the BabA adhesin and to a lesser extent with the SabA adhesin. This agrees with reports, that MUC5B carries both fucosylated blood group antigens and alpha2-3-linked sialic acids. The adhesion of H. pylori to fibronectin and lactoferrin persisted in the babA/sabA double mutant. Because binding to these molecules was abolished by denaturation rather than by deglycosylation, it was suggested to depend on the recognition of unknown receptor moieties by an additional unknown bacterial surface component. The results demonstrate that the bacterial overlay method on glycoconjugate arrays is a useful tool for exploration and the characterization of unknown adhesin specificities of H. pylori and other bacteria.


Assuntos
Aderência Bacteriana , Glicoconjugados/metabolismo , Helicobacter pylori/fisiologia , Adesinas Bacterianas/metabolismo , Sequência de Carboidratos , Eletroforese em Gel de Poliacrilamida , Fibronectinas/metabolismo , Glicoconjugados/química , Helicobacter pylori/genética , Lactoferrina/metabolismo , Dados de Sequência Molecular
13.
Int J Med Microbiol ; 292(3-4): 247-56, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12398215

RESUMO

The attachment of the bacterial pathogen Helicobacter pylori (H. pylori) to gastric epithelial cells is commonly believed to be required for an efficient and persistent colonization of the human stomach as well as for host cell trans-membrane signaling. In the past, several putative adhesins were postulated, including the outer membrane proteins AlpAB and the bacterial lipopolysaccharide (LPS) presenting oligomeric Lewis x (Le(x)) sugar components. We investigated the AlpAB-mediated and the Le(x)-dependent binding by knockout mutagenesis in one distinct strain, H. pylori P1. We show here that the mutagenesis of either alpA and/or alpB dramatically reduced the adherence of H. pylori P1 to a given gastric biopsy section. None of these mutations influenced the surface exposure of the Le(x) antigen, suggesting that the assembly and/or presentation of LPS is independent of the AlpAB outer membrane proteins. However, a truncation of the LPS O-side chain by a galE mutation abolished the presentation of the Le(x) epitope. This Le(x)-negative strain did not show any significant reduction in its binding capacity to the gastric tissue, when compared with the corresponding wild-type strain. From these data we conclude that the AlpAB-specific adherence is independent of the composition of the LPS and that the oligomeric Le(x) structure does not confer binding to the gastric biopsy material used in this study. As the adhesion properties of our H. pylori strain P1 vary in dependence on the respective biopsy donor it is assumed that the surface-exposed Le(x) epitope recognizes a different host cell receptor than AlpAB, which was probably not present in the tissue sections used in this study.


Assuntos
Adesinas Bacterianas/fisiologia , Aderência Bacteriana/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/fisiologia , Gastropatias/microbiologia , Adesinas Bacterianas/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Western Blotting , DNA Bacteriano/química , DNA Bacteriano/genética , Imunofluorescência , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/metabolismo , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Humanos , Técnicas In Vitro , Antígenos CD15/metabolismo , Mutagênese , Reação em Cadeia da Polimerase , Gastropatias/metabolismo
14.
Int J Med Microbiol ; 292(3-4): 257-66, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12398216

RESUMO

In several studies Helicobacter pylori type I strains (cag-positive strains) have been described to translocate their CagA protein into epithelial cells, where it is tyrosine-phosphorylated. The intimate contact allows a Cag-dependent bacteria-to-cell signaling inducing the secretion of the chemokine interleukin-8. Although a contact between the bacterial and the eukaryotic cell is known to be necessary for these signal transduction events the bacterial adhesin and the cellular receptor are unknown, so far. In this study, we investigated the influence of several outer membrane proteins associated with adherence on CagA translocation and IL-8 induction. The quantitative assessment of a cag deletion mutant strain binding to epithelial cells revealed that the Cag secretion apparatus is not primarily necessary for attachment. In contrast, the knockout mutation of the adherence-associated alpAB locus significantly reduced the binding capacity in two independent strains. Despite this partial adherence defect, the alpAB mutation did not affect CagA translocation and IL-8 induction. The mutagenesis of the bab group genes hp317, hp896 and hp1243 in H. pylori 26695 did not influence the Cag-dependent signaling either. No causative linkage could be found between the production of the outer membrane proteins HopZ, OipA or seven additional outer membrane proteins and CagA translocation or IL-8 induction.


Assuntos
Antígenos de Bactérias , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/metabolismo , Helicobacter pylori/metabolismo , Interleucina-8/biossíntese , Adesinas Bacterianas/metabolismo , Aderência Bacteriana/fisiologia , Proteínas da Membrana Bacteriana Externa/genética , Western Blotting , DNA Bacteriano/química , DNA Bacteriano/genética , Helicobacter pylori/genética , Interleucina-8/metabolismo , Mutagênese Insercional , Fosforilação , Transdução de Sinais/fisiologia , Tirosina/metabolismo
15.
Microbiology (Reading) ; 144 ( Pt 9): 2505-2516, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9782498

RESUMO

The function of the pfr gene encoding the ferritin from Helicobacter pylori was investigated using the Fur titration assay (FURTA) in Escherichia coli, and by characterization of a pfr-deficient mutant strain of H. pylori. Nucleotide sequence analysis revealed that the pfr region is conserved among strains (> 95% nucleotide identity). Two transcriptional start sites, at least one of them preceded by a sigma 70-dependent promoter, were identified. Provision of the H. pylori pfr gene on a multicopy plasmid resulted in reversal of the Fur-mediated repression of the fhuF gene in E. coli, thus enabling the use of the FURTA for cloning of the ferritin gene. Inactivation of the pfr gene, either by insertion of a resistance cassette or by deletion of the up- and downstream segments, abolished this function. Immunoblot analysis with a Pfr-specific antiserum detected the Pfr protein in H. pylori and in E. coli carrying the pfr gene on a plasmid. Pfr-deficient mutants of H. pylori were generated by marker-exchange mutagenesis. These were more susceptible than the parental strain to killing by various metal ions including irons, copper and manganese, whereas conditions of oxidative stress or iron deprivation were not discriminative. Analysis by element-specific electron microscopy revealed that growth of H. pylori in the presence of iron induces the formation of two kinds of cytoplasmic aggregates: large vacuole-like bodies and smaller granules containing iron in association with oxygen or phosphorus. Neither of these structures was detected in the pfr-deficient mutant strain. Furthermore, the ferritin accumulated under iron overload and the pfr-deficient mutant strains lacked expression of a 12 kDa protein which was negatively regulated by iron in the parental strain. The results indicate that the nonhaem-iron ferritin is involved in the formation of iron-containing subcellular structures and contributes to metal resistance of H. pylori. Further evidence for an interaction of ferritin with iron-dependent regulation mechanisms is provided.


Assuntos
Ferritinas/genética , Genes Bacterianos , Helicobacter pylori/genética , Sequência de Aminoácidos , Sequência de Bases , Clonagem Molecular , Primers do DNA/genética , DNA Bacteriano/genética , Escherichia coli/genética , Ferritinas/biossíntese , Ferritinas/isolamento & purificação , Helicobacter pylori/metabolismo , Helicobacter pylori/ultraestrutura , Ferro/metabolismo , Microscopia Eletrônica , Dados de Sequência Molecular , Mutação , RNA Bacteriano/genética , Transcrição Gênica , Vacúolos/metabolismo , Vacúolos/ultraestrutura
16.
J Clin Microbiol ; 40(6): 2244-6, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12037098

RESUMO

The BabA, cagA, and vacA statuses of 827 Helicobacter pylori isolates were used in logistic regression models to discriminate duodenal ulcer from gastritis. Only BabA was a candidate for a universal virulence factor, but the low c statistic value (0.581) indicates that none of these factors were helpful in predicting the clinical presentation.


Assuntos
Adesinas Bacterianas , Antígenos de Bactérias , Úlcera Duodenal/diagnóstico , Úlcera Duodenal/microbiologia , Gastrite/diagnóstico , Infecções por Helicobacter/complicações , Helicobacter pylori/patogenicidade , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Gastrite/microbiologia , Infecções por Helicobacter/microbiologia , Humanos , Modelos Logísticos , Virulência
18.
Biochemistry ; 43(11): 3089-103, 2004 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-15023060

RESUMO

Helicobacter pylori produces one monofunctional catalase, encoded by katA (hp0875). The crystal structure of H. pylori catalase (HPC) has been determined and refined at 1.6 A with crystallographic agreement factors R and R(free) of 17.4 and 21.9%, respectively. The crystal exhibits P2(1)2(1)2 space group symmetry and contains two protein subunits in the asymmetric unit. The core structure of the HPC subunit, including the disposition of a heme b prosthetic group, is closely related to those of other catalases, although it appears to be the only clade III catalase that has been characterized that does not bind NADPH. The heme iron in one subunit of the native enzyme appears to be covalently modified, possibly with a perhydroxy or dioxygen group in a compound III-like structure. Formic acid is known to bind in the active site of catalases, promoting the breakdown of reaction intermediates compound I and compound II. The structure of an HPC crystal soaked with sodium formate at pH 5.6 has also been determined to 1.6 A (with R and R(free) values of 18.1 and 20.7%, respectively), revealing at least 36 separate formate or formic acid residues in the HPC dimer. In turn, the number of water molecules refined into the models decreased from 1016 in the native enzyme to 938 in the formate-treated enzyme. Extra density, interpreted as azide, is found in a location of both structures that involves interaction with all four subunits in the tetramer. Electron paramagnetic resonance spectra confirm that azide does not bind as a ligand of the iron and that formate does bind in the heme pocket. The stability of the formate or formic acid molecule found inside the heme distal pocket has been investigated by calculations based on density functional theory.


Assuntos
Proteínas de Bactérias/química , Catalase/química , Formiatos/química , Helicobacter pylori/enzimologia , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/antagonistas & inibidores , Sítios de Ligação , Catalase/antagonistas & inibidores , Bovinos , Simulação por Computador , Cristalografia por Raios X , Dimerização , Espectroscopia de Ressonância de Spin Eletrônica , Inibidores Enzimáticos/química , Estabilidade Enzimática , Heme/química , Humanos , Modelos Químicos , Modelos Moleculares , Dados de Sequência Molecular , Subunidades Proteicas/química
19.
Infect Immun ; 70(7): 3923-9, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12065536

RESUMO

The reactivity of the essential element iron necessitates a concerted expression of ferritins, which mediate iron storage in a nonreactive state. Here we have further established the role of the Helicobacter pylori ferritin Pfr in iron metabolism and gastric colonization. Iron stored in Pfr enabled H. pylori to multiply under severe iron starvation and protected the bacteria from acid-amplified iron toxicity, as inactivation of the pfr gene restricted growth of H. pylori under these conditions. The lowered total iron content in the pfr mutant, which is probably caused by decreased iron uptake rates, was also reflected by an increased resistance to superoxide stress. Iron induction of Pfr synthesis was clearly diminished in an H. pylori feoB mutant, which lacked high-affinity ferrous iron transport, confirming that Pfr expression is mediated by changes in the cytoplasmic iron pool and not by extracellular iron. This is well in agreement with the recent discovery that iron induces Pfr synthesis by abolishing Fur-mediated repression of pfr transcription, which was further confirmed here by the observation that iron inhibited the in vitro binding of recombinant H. pylori Fur to the pfr promoter region. The functions of H. pylori Pfr in iron metabolism are essential for survival in the gastric mucosa, as the pfr mutant was unable to colonize in a Mongolian gerbil-based animal model. In summary, the pfr phenotypes observed give new insights into prokaryotic ferritin functions and indicate that iron storage and homeostasis are of extraordinary importance for H. pylori to survive in its hostile natural environment.


Assuntos
Proteínas de Bactérias/fisiologia , Ferritinas/análogos & derivados , Ferritinas/fisiologia , Helicobacter pylori/metabolismo , Ferro/metabolismo , Estômago/microbiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Transporte Biológico , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Modelos Animais de Doenças , Ferritinas/genética , Gerbillinae , Infecções por Helicobacter/genética , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Helicobacter pylori/crescimento & desenvolvimento , Concentração de Íons de Hidrogênio , Mutagênese , Estresse Oxidativo , Paraquat/farmacologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Superóxidos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA