Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Sci Rep ; 14(1): 7683, 2024 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-38561502

RESUMO

Helicobacter pylori (H. pylori), known for causing gastric inflammation, gastritis and gastric cancer, prompted our study to investigate the differential expression of cytokines in gastric tissues, which is crucial for understanding H. pylori infection and its potential progression to gastric cancer. Focusing on Il-1ß, IL-6, IL-8, IL-12, IL-18, and TNF-α, we analysed gene and protein levels to differentiate between H. pylori-infected and non-infected gastritis. We utilised real-time quantitative polymerase chain reaction (RT-qPCR) for gene quantification, immunohistochemical staining, and ELISA for protein measurement. Gastric samples from patients with gastritis were divided into three groups: (1) non-gastritis (N-group) group, (2) gastritis without H. pylori infection (G-group), and (3) gastritis with H. pylori infection (GH-group), each consisting of 8 samples. Our findings revealed a statistically significant variation in cytokine expression. Generally, cytokine levels were higher in gastritis, but in H. pylori-infected gastritis, IL-1ß, IL-6, and IL-8 levels were lower compared to H. pylori-independent gastritis, while IL-12, IL-18, and TNF-α levels were higher. This distinct cytokine expression pattern in H. pylori-infected gastritis underscores a unique inflammatory response, providing deeper insights into its pathogenesis.


Assuntos
Gastrite , Infecções por Helicobacter , Helicobacter pylori , Helicobacter , Neoplasias Gástricas , Humanos , Citocinas/metabolismo , Helicobacter pylori/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Helicobacter/metabolismo , Interleucina-8/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Gastrite/patologia , Interleucina-12/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Infecções por Helicobacter/genética , Infecções por Helicobacter/metabolismo , Mucosa Gástrica/metabolismo
2.
Apoptosis ; 29(3-4): 439-456, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38001345

RESUMO

Gastric cancer is strongly associated with Helicobacter pylori (H. pylori) infection. However, the molecular mechanisms underlying the development of gastric cancer in the context of H. pylori infection, particularly in relation to ferroptosis, remain poorly understood. In this study, we investigated the role of the Helicobacter-associated ferroptosis gene YWHAE in gastric cancer. We analyzed multi-omics data, performed molecular docking, and employed machine learning to comprehensively evaluate the expression, function, and potential implications in gastric cancer, including its influence on drug sensitivity, mutation, immune microenvironment, immunotherapy, and prognosis. Our findings demonstrated that the YWHAE gene exhibits high expression in both H. pylori-associated gastritis and gastric cancer. Pan-cancer analysis revealed elevated expression of YWHAE in several cancer types compared to normal tissues. We also examined the methylation, single nucleotide variations (SNVs), and copy number variations (CNVs) associated with YWHAE. Single-cell analysis indicated that the YWHAE gene is expressed in various cell types, with its expression level potentially influenced by H. pylori infection. Functionally, we observed a positive correlation between YWHAE gene expression and ferroptosis in gastric cancer and associated with multiple cancer-related signaling pathways, including MAPK, NF-κB, and PI3K. Furthermore, we predicted five small molecule compounds that show promise for treating gastric cancer patients and screened five drugs with the highest correlation with YWHAE and validated them by molecular docking. Additionally, significant differences were observed in various immune cell types and immunotherapeutic response between the high and low YWHAE gene expression groups. Moreover, we found a positive correlation between YWHAE gene expression and the tumour mutation burden (TMB). By applying 10 machine learning algorithms and 101 integration combinations, we developed a prognostic model for YWHAE-related genes. Finally, qRT-PCR and immunohistochemistry (IHC) consistently demonstrated the upregulation of YWHAE in gastric cancer. In conclusion, we conducted a comprehensive analysis of YWHAE gene in gastric cancer. Our findings provided novel insights into the role of YWHAE as a gene associated with H. pylori infection and ferroptosis in gastric cancer and expanded our understanding of the molecular mechanisms underlying gastric carcinogenesis.


Assuntos
Ferroptose , Helicobacter pylori , Helicobacter , Neoplasias Gástricas , Humanos , Helicobacter/metabolismo , Simulação de Acoplamento Molecular , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Variações do Número de Cópias de DNA , Ferroptose/genética , Multiômica , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Apoptose , Microambiente Tumoral , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo
3.
Immunol Cell Biol ; 101(5): 444-457, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36967659

RESUMO

Helicobacter pylori (H. pylori) infection can trigger chronic gastric inflammation perpetuated by overactivation of the innate immune system, leading to a cascade of precancerous lesions culminating in gastric cancer. However, key regulators of innate immunity that promote H. pylori-induced gastric pathology remain ill-defined. The innate immune cytosolic DNA sensor absent in melanoma 2 (AIM2) contributes to the pathogenesis of numerous autoimmune and chronic inflammatory diseases, as well as cancers including gastric cancer. We therefore investigated whether AIM2 contributed to the pathogenesis of Helicobacter-induced gastric disease. Here, we reveal that AIM2 messenger RNA and protein expression levels are elevated in H. pylori-positive versus H. pylori-negative human gastric biopsies. Similarly, chronic Helicobacter felis infection in wild-type mice augmented Aim2 gene expression levels compared with uninfected controls. Notably, gastric inflammation and hyperplasia were less severe in H. felis-infected Aim2-/- versus wild-type mice, evidenced by reductions in gastric immune cell infiltrates, mucosal thickness and proinflammatory cytokine and chemokine release. In addition, H. felis-driven proliferation and apoptosis in both gastric epithelial and immune cells were largely attenuated in Aim2-/- stomachs. These observations in Aim2-/- mouse stomachs correlated with decreased levels of inflammasome activity (caspase-1 cleavage) and the mature inflammasome effector cytokine, interleukin-1ß. Taken together, this work uncovers a pathogenic role for the AIM2 inflammasome in Helicobacter-induced gastric disease, and furthers our understanding of the host immune response to a common pathogen and the complex and varying roles of AIM2 at different stages of cancerous and precancerous gastric disease.


Assuntos
Felis , Helicobacter , Lesões Pré-Cancerosas , Neoplasias Gástricas , Animais , Humanos , Camundongos , Citocinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Felis/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Helicobacter/metabolismo , Inflamassomos/metabolismo , Inflamação/patologia , Lesões Pré-Cancerosas/patologia
4.
Metallomics ; 14(9)2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-36002005

RESUMO

Acquisition and homeostasis of essential metals during host colonization by bacterial pathogens rely on metal uptake, trafficking, and storage proteins. How these factors have evolved within bacterial pathogens is poorly defined. Urease, a nickel enzyme, is essential for Helicobacter pylori to colonize the acidic stomach. Our previous data suggest that acquisition of nickel transporters and a histidine-rich protein (HRP) involved in nickel storage in H. pylori and gastric Helicobacter spp. have been essential evolutionary events for gastric colonization. Using bioinformatics, proteomics, and phylogenetics, we extended this analysis to determine how evolution has framed the repertoire of HRPs among 39 Epsilonproteobacteria; 18 gastric and 11 non-gastric enterohepatic (EH) Helicobacter spp., as well as 10 other Epsilonproteobacteria. We identified a total of 213 HRPs distributed in 22 protein families named orthologous groups (OGs) with His-rich domains, including 15 newly described OGs. Gastric Helicobacter spp. are enriched in HRPs (7.7 ± 1.9 HRPs/strain) as compared to EH Helicobacter spp. (1.9 ± 1.0 HRPs/strain) with a particular prevalence of HRPs with C-terminal histidine-rich domains in gastric species. The expression and nickel-binding capacity of several HRPs was validated in five gastric Helicobacter spp. We established the evolutionary history of new HRP families, such as the periplasmic HP0721-like proteins and the HugZ-type heme oxygenases. The expansion of histidine-rich extensions in gastric Helicobacter spp. proteins is intriguing but can tentatively be associated with the presence of the urease nickel enzyme. We conclude that this HRP expansion is associated with unique properties of organisms that rely on large intracellular nickel amounts for their survival.


Assuntos
Helicobacter pylori , Helicobacter , Proteínas de Bactérias/metabolismo , Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Histidina/metabolismo , Níquel/metabolismo , Proteínas , Estômago , Urease/metabolismo
5.
Vet Res ; 53(1): 42, 2022 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-35692057

RESUMO

This article focuses on the pathogenic significance of Helicobacter species naturally colonizing the stomach of dogs, cats and pigs. These gastric "non-Helicobacter (H.) pylori Helicobacter species" (NHPH) are less well-known than the human adapted H. pylori. Helicobacter suis has been associated with gastritis and decreased daily weight gain in pigs. Several studies also attribute a role to this pathogen in the development of hyperkeratosis and ulceration of the non-glandular stratified squamous epithelium of the pars oesophagea of the porcine stomach. The stomach of dogs and cats can be colonized by several Helicobacter species but their pathogenic significance for these animals is probably low. Helicobacter suis as well as several canine and feline gastric Helicobacter species may also infect humans, resulting in gastritis, peptic and duodenal ulcers, and low-grade mucosa-associated lymphoid tissue lymphoma. These agents may be transmitted to humans most likely through direct or indirect contact with dogs, cats and pigs. Additional possible transmission routes include consumption of water and, for H. suis, also consumption of contaminated pork. It has been described that standard H. pylori eradication therapy is usually also effective to eradicate the NHPH in human patients, although acquired antimicrobial resistance may occasionally occur and porcine H. suis strains are intrinsically less susceptible to aminopenicillins than non-human primate H. suis strains and other gastric Helicobacter species. Virulence factors of H. suis and the canine and feline gastric Helicobacter species include urease activity, motility, chemotaxis, adhesins and gamma-glutamyl transpeptidase. These NHPH, however, lack orthologs of cytotoxin-associated gene pathogenicity island and vacuolating cytotoxin A, which are major virulence factors in H. pylori. It can be concluded that besides H. pylori, gastric Helicobacter species associated with dogs, cats and pigs are also clinically relevant in humans. Although recent research has provided better insights regarding pathogenic mechanisms and treatment strategies, a lot remains to be investigated, including true prevalence rates, exact modes of transmission and molecular pathways underlying disease development and progression.


Assuntos
Doenças do Gato , Doenças do Cão , Gastrite , Infecções por Helicobacter , Helicobacter heilmannii , Helicobacter pylori , Helicobacter , Doenças dos Suínos , Animais , Gatos , Citotoxinas , Cães , Mucosa Gástrica/metabolismo , Gastrite/veterinária , Helicobacter/metabolismo , Infecções por Helicobacter/patologia , Infecções por Helicobacter/veterinária , Helicobacter heilmannii/genética , Helicobacter pylori/metabolismo , Humanos , Suínos , Fatores de Virulência/genética
6.
Photosynth Res ; 143(3): 241-250, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31838634

RESUMO

The heliobacterial reaction center (HbRC) is the simplest known photochemical reaction center, in terms of its polypeptide composition. In the heliobacterial cells, its electron donor is a cytochrome (cyt) c553 attached to the membrane via a covalent linkage with a diacylglycerol. We have reconstituted purified HbRC into liposomes mimicking the phospholipid composition of heliobacterial membranes. We also incorporated a lipid with a headgroup containing Ni(II):nitrilotriacetate (NTA) to provide a binding site for the soluble version of the heliobacterial cyt c553 in which the N-terminal membrane attachment site is replaced by a hexahistidine tag. The HbRC was inserted into the liposomes with the donor side preferentially exposed to the exterior; this bias increased to nearly 100% with higher concentrations (≥ 10 mol%) of the Ni(II)-NTA lipid in the membrane, and is most likely due to the net negative charge of the surface of the membrane. The HbRC in proteoliposomes without the Ni(II)-NTA lipid exhibited normal charge separation and subsequent charge recombination of the P800+FX- state in 15 ms; however, the oxidized primary donor (P800+) was not significantly reduced by added H6-cyt c553. In contrast, with proteoliposomes containing the Ni(II)-NTA lipid, addition of H6-cyt c553 resulted in a new kinetic component resulting from fast reduction (2-5 ms) of P800+ by H6-cyt c553. The contribution of this kinetic component varied with the concentration of added H6-cyt c553 and could represent 80% or more of the total P800+ decay. Thus, the HbRC and its interaction with its native electron donor have been reconstituted into an artificial membrane system.


Assuntos
Grupo dos Citocromos c/metabolismo , Helicobacter/metabolismo , Processos Fotoquímicos , Complexo de Proteínas do Centro de Reação Fotossintética/metabolismo , Proteolipídeos/metabolismo , Transporte de Elétrons , Flavodoxina/metabolismo , Oxirredução , Fatores de Tempo
7.
Sci Rep ; 8(1): 15453, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30337679

RESUMO

Helicobacter (H.) pylori is an important risk factor for gastric malignancies worldwide. Its outer membrane proteome takes an important role in colonization of the human gastric mucosa. However, in zoonotic non-H. pylori helicobacters (NHPHs) also associated with human gastric disease, the composition of the outer membrane (OM) proteome and its relative contribution to disease remain largely unknown. By means of a comprehensive survey of the diversity and distribution of predicted outer membrane proteins (OMPs) identified in all known gastric Helicobacter species with fully annotated genome sequences, we found genus- and species-specific families known or thought to be implicated in virulence. Hop adhesins, part of the Helicobacter-specific family 13 (Hop, Hor and Hom) were restricted to the gastric species H. pylori, H. cetorum and H. acinonychis. Hof proteins (family 33) were putative adhesins with predicted Occ- or MOMP-family like 18-stranded ß-barrels. They were found to be widespread amongst all gastric Helicobacter species only sporadically detected in enterohepatic Helicobacter species. These latter are other members within the genus Helicobacter, although ecologically and genetically distinct. LpxR, a lipopolysaccharide remodeling factor, was also detected in all gastric Helicobacter species but lacking as well from the enterohepatic species H. cinaedi, H. equorum and H. hepaticus. In conclusion, our systemic survey of Helicobacter OMPs points to species and infection-site specific members that are interesting candidates for future virulence and colonization studies.


Assuntos
Proteínas da Membrana Bacteriana Externa/genética , Simulação por Computador , Helicobacter/genética , Filogenia , Proteômica , Proteínas da Membrana Bacteriana Externa/metabolismo , Helicobacter/metabolismo
8.
Sci Rep ; 7(1): 9909, 2017 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-28855660

RESUMO

Helicobacter pullorum is an avian bacterium that causes gastroenteritis, intestinal bowel and hepatobiliary diseases in humans. Although H. pullorum has been shown to activate the mammalian innate immunity with release of nitric oxide (NO), the proteins that afford protection against NO and reactive nitrogen species (RNS) remain unknown. Here several protein candidates of H. pullorum, namely a truncated (TrHb) and a single domain haemoglobin (SdHb), and three peroxiredoxin-like proteins (Prx1, Prx2 and Prx3) were investigated. We report that the two haemoglobin genes are induced by RNS, and that SdHb confers resistance to nitrosative stress both in vitro and in macrophages. For peroxiredoxins, the prx2 and prx3 expression is enhanced by peroxynitrite and hydrogen peroxide, respectively. Mutation of prx1 does not alter the resistance to these stresses, while the single ∆prx2 and double ∆prx1∆prx2 mutants have decreased viability. To corroborate the physiological data, the biochemical analysis of the five recombinant enzymes was done, namely by stopped-flow spectrophotometry. It is shown that H. pullorum SdHb reacts with NO much more quickly than TrHb, and that the three Prxs react promptly with peroxynitrite, Prx3 displaying the highest reactivity. Altogether, the results unveil SdHb and Prx3 as major protective systems of H. pullorum against nitrosative stress.


Assuntos
Infecções por Helicobacter/microbiologia , Helicobacter/patogenicidade , Estresse Nitrosativo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Helicobacter/genética , Helicobacter/metabolismo , Infecções por Helicobacter/patologia , Humanos , Intestinos/microbiologia , Intestinos/patologia , Fígado/microbiologia , Fígado/patologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Viabilidade Microbiana/genética , Mutação , Óxido Nítrico/metabolismo , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Virulência
9.
J Proteomics ; 97: 245-55, 2014 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23899588

RESUMO

Helicobacter pylori is a major human pathogen that infects the gastric mucosa and is responsible for a range of infections including gastritis and gastric carcinoma. Although other bacteria within the Helicobacter genus can also infect the gastric mucosa, there are Helicobacter species that infect alternative sites within the gastrointestinal (GI) tract. Two-dimensional gel electrophoresis was used to compare the cellular proteomes of seven non-pylori Helicobacters (H. mustelae, H. felis, H. cinaedi, H. hepaticus, H. fennelliae, H. bilis and H. cholecystus) against the more extensively characterised H. pylori. The different Helicobacter species showed distinctive 2D protein profiles, it was possible to combine them into a single dataset using Progenesis SameSpots software. Principal Component Analysis was used to search for correlations between the bacterial proteomes and their sites of infection. This approach clearly discriminated between gastric (i.e. those which infect in the gastric mucosa) and enterohepatic Helicobacter species (i.e. those bacteria that infect the small intestine and hepatobillary regions of the GI tract). Selected protein spots showing significant differences in abundance between these two groups of bacteria were identified by LC-MS. The data provide an initial insight into defining those features of the bacterial proteome that influence the sites of bacterial infection. BIOLOGICAL SIGNIFICANCE: This study demonstrated that representative members of the Helicobacter genus were readily discriminated from each other on the basis of their in vitro whole cell proteomes determined using 2D gel electrophoresis. Despite the intra-species heterogeneity observed it was possible, to demonstrate that the enterohepatic (represented by H. bilis, H. hepaticus, H. fennelliae, H. cinaedi and H. cholecystus) and gastric (represented by H. pylori, H. mustelae, and H. felis) Helicobacters formed discrete groups based on their 2D protein profiles. A provisional proteomic signature was identified that correlated with the typical sites of colonisation of these members of the Helicobacter genus. This article is part of a Special Issue entitled: Trends in Microbial Proteomics.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter/metabolismo , Proteoma/metabolismo , Proteômica , Proteínas de Bactérias/genética , Eletroforese em Gel Bidimensional , Helicobacter/genética , Infecções por Helicobacter/genética , Humanos , Especificidade da Espécie
10.
Vet Res ; 44: 56, 2013 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-23865636

RESUMO

Genomic analysis of a metronidazole resistant H. bizzozeronii strain revealed a frame length extension of the oxygen-insensitive NAD(P)H-nitroreductase HBZC1_00960 (RdxA), associated with the disruption of the C-terminal cysteine-containing conserved region (IACLXALGK). This was the result of the extension (from C8 to C9) of a simple sequence cytosine repeat (SSCR) located in the 3' of the gene. A 3' SSCR is also present in the rdxA homolog of H. heilmannii sensu stricto, but not in H. pylori. We showed that in the majority of in vitro spontaneous H. bizzozeronii metronidazole resistant mutants, the extension of the 3' SSCR of rdxA was the only mutation observed. In addition, we observed that H. bizzozeronii ΔrdxA mutant strain showed the same MIC value of metronidazole observed in the spontaneous mutants. These data indicate that loss of function mutations in rdxA and in particular the disruption of the conserved region IACLXALGK is associated with reduced susceptibility to metronidazole in H. bizzozeronii. Slipped-strand mispairing of the SSCR located in the 3' of the H. bizzozeronii rdxA appears to be the main mechanism. We also observed that H. bizzozeronii acquires resistance to metronidazole at high mutation rate, and that serial passages in vitro without selection induced an increased level of susceptibility. In conclusion, contrary to what was previously described in H. pylori, the H. bizzozeronii rdxA appears to be a contingency gene which undergoes phase variation. The contingency nature of rdxA should be carefully considered when metronidazole is used in the treatment of H. heilmannii-associated gastritis.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Farmacorresistência Bacteriana , Helicobacter/efeitos dos fármacos , Helicobacter/genética , Metronidazol/farmacologia , Nitrorredutases/genética , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Cães , Helicobacter/metabolismo , Humanos , Mutação , Nitrorredutases/química , Nitrorredutases/metabolismo , Alinhamento de Sequência/veterinária
11.
Asian Pac J Cancer Prev ; 12(12): 3421-5, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22471491

RESUMO

AIM: The potential role of GSTM1, GSTT1 and GSTP1 polymorphisms in risk of gastric cancer in Chinese was studied. METHODS: We collected 194 gastric cancers by pathologic examination and 412 controls from southern China during January 2007 to January 2011. Genotyping was based upon duplex polymerase-chain-reaction with the PCR-CTPP method. RESULTS: Individuals carrying null GSTM1 and GSTT1 had 1.49 and 1.96 fold risk sof gastric cancer when compared with respective non-null genotypes. We also found a non-significant 37% excess risk of gastric cancer among carriers of GSTP1 1b/1b genotype when compared with 1a/1a genotype (OR=1.37, 95% CI=0.81-2.25). The combination of null/null GSTM1 and GSTT1 genotypes showed higher increased risk of gastric cancer (OR=3.17, 95% CI=1.68-4.21). Moreover, cancers in ever smokers and ever drinkers were observed to be strongly associated with null GSTM1 and GSTT1, and a significant cancer risk was observed in positive H.pylori infection individuals with null GSTT1. CONCLUSION: Our study provided evidence that genetic deletion of GSTM1 and GSTT1 may contribute to increased susceptibility to gastric cancer in our Chinese population, while the GSTP1a/b polymorphism may not.


Assuntos
Povo Asiático/genética , Glutationa S-Transferase pi/genética , Glutationa Transferase/genética , Polimorfismo Genético/genética , Neoplasias Gástricas/genética , Adulto , Idoso , Consumo de Bebidas Alcoólicas , Estudos de Casos e Controles , DNA/genética , Suscetibilidade a Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Seguimentos , Genótipo , Helicobacter/metabolismo , Infecções por Helicobacter/genética , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/virologia , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Prognóstico , Fatores de Risco , Fumar , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/virologia
12.
Mol Microbiol ; 73(6): 1058-71, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19682263

RESUMO

Cytochromes of the c-type function on the outer side of the cytoplasmic membrane in bacteria where they also are assembled from apo-cytochrome polypeptide and haem. Two distinctly different systems for cytochrome c maturation are found in bacteria. System I present in Escherichia coli has eight to nine different Ccm proteins. System II is found in Bacillus subtilis and comprises four proteins: CcdA, ResA, ResB and ResC. ResB and ResC are poorly understood polytopic membrane proteins required for cytochrome c synthesis. We have analysed these two B. subtilis proteins produced in E. coli and in the native organism. ResB is shown to bind protohaem IX and haem is found covalently bound to residue Cys-138. Results in B. subtilis suggest that also ResC can bind haem. Our results complement recent findings made with Helicobacter CcsBA supporting the hypothesis that ResBC as a complex translocates haem by attaching it to ResB on the cytoplasmic side of the membrane and then transferring it to an extra-cytoplasmic location in ResC, from where it is made available to the apo-cytochromes.


Assuntos
Bacillus subtilis/enzimologia , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Citocromos c/metabolismo , Heme/metabolismo , Hemeproteínas/metabolismo , Sequência de Aminoácidos , Bacillus subtilis/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Helicobacter/enzimologia , Helicobacter/metabolismo , Proteínas Ligantes de Grupo Heme , Dados de Sequência Molecular , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
13.
Ann N Y Acad Sci ; 1125: 242-51, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18378596

RESUMO

Anaerobic microorganisms (such as clostridia) present in the large intestine of animals generate molecular hydrogen (H(2)) by fermentation using "H(2)-evolving" hydrogenases. The gas can also be detected in other tissues in mice, including the stomach, liver, spleen, or small intestine. It is established that this available H(2) can in turn be used as a source of energy by some pathogenic bacteria, including Helicobacter species like H. pylori and H. hepaticus. Both species possess one hydrogenase, which has been studied for H(2) oxidation characteristics and for its role in conferring animal colonization. On the basis of available annotated gene sequences, other Helicobacter species also appear to have one well-conserved respiratory, membrane-bound, nickel-iron-containing [NiFe] hydrogenase. Although H. pylori has been well-studied, many other (poorly studied) Helicobacter species likely represent a spectrum of emerging pathogens. The important role of hydrogenases in Helicobacter species is discussed, and the hydrogenases, their maturation/accessory factors, their regulation, as well as nickel transport and metabolism among the different species are compared.


Assuntos
Helicobacter/metabolismo , Hidrogênio/metabolismo , Níquel/metabolismo , Proteínas de Bactérias/metabolismo , Helicobacter/enzimologia , Helicobacter/patogenicidade , Hidrogenase/metabolismo , Cinética , Oxirredução
14.
J Clin Microbiol ; 44(5): 1650-8, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16672389

RESUMO

The growth of the gastric pathogen Helicobacter pylori in the absence of serum remains challenging, and nutritional requirements have only partially been defined, while almost nothing is known about nutritional requirements of other Helicobacter spp. Although previous data showed that H. pylori grows in the chemically defined medium F-12, but not in other tissue culture media examined, the specific components responsible for growth were not entirely understood. Here we describe the optimization of amino acids, metals, and sodium chloride for H. pylori. Iron, zinc, and magnesium were critical for growth; copper was not required. Optimization of sodium chloride was further beneficial. Nutritional requirements and antibiotic resistance patterns of several other Helicobacter spp. revealed that all except H. felis grew in serum-free, unsupplemented F-12. All Helicobacter spp. were resistant to at least six antimicrobial agents when cultured in the presence of serum. However, in the absence of serum, H. pylori, H. mustelae, and H. muridarum became sensitive to polymyxin B and/or trimethoprim. Much of the data were obtained using a convenient ATP assay to quantify growth. H. pylori has surprisingly few absolute requirements for growth: 9 amino acids, sodium and potassium chloride, thiamine, iron, zinc, magnesium, hypoxanthine, and pyruvate. These data suggest that H. pylori and other Helicobacter spp. are not as fastidious as previously thought. The data also suggest that chemically defined media described herein could yield the growth of a wide range of Helicobacter spp., allowing a more detailed characterization of Helicobacter physiology and interactions with host cells.


Assuntos
Helicobacter/efeitos dos fármacos , Helicobacter/metabolismo , Trifosfato de Adenosina/metabolismo , Aminoácidos/metabolismo , Contagem de Colônia Microbiana/métodos , Meios de Cultura Livres de Soro/química , Farmacorresistência Bacteriana , Helicobacter/crescimento & desenvolvimento , Ferro/metabolismo , Metais/metabolismo , Sais/metabolismo , Cloreto de Sódio/metabolismo , Vitaminas/metabolismo
15.
Biochemistry ; 45(20): 6306-16, 2006 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-16700542

RESUMO

Electron transfer in the membranes and the type I reaction center (RC) core protein complex isolated from Heliobacterium modesticaldum was studied by optical and ESR spectroscopy. The RC is a homodimer of PshA proteins. In the isolated membranes, illumination at 14 K led to accumulation of a stable ESR signal of the reduced iron-sulfur center F(B)(-) in the presence of dithiothreitol, and an additional 20 min illumination at 230 K induced the spin-interacting F(A)(-)/F(B)(-) signal at 14 K. During illumination at 5 K in the presence of dithionite, we detected a new transient signal with the following values: g(z)= 2.040, g(y)= 1.911, and g(x)= 1.896. The signal decayed rapidly with a 10 ms time constant after the flash excitation at 5 K and was attributed to the F(X)(-)-type center, although the signal shape was more symmetrical than that of F(X)(-) in photosystem I. In the purified RC core protein, laser excitation induced the absorption change of a special pair, P800. The flash-induced P800(+) signal recovered with a fast 2-5 ms time constant below 150 K, suggesting charge recombination with F(X)(-). Partial destruction of the RC core protein complex by a brief exposure to air increased the level of the P800(+)A(0)(-) state that gave a lifetime (t(1/2)) of 100 ns at 77 K. The reactions of F(X) and quinone were discussed on the basis of the three-dimensional structural model of RC that predicts the conserved F(X)-binding site and the quinone-binding site, which is more hydrophilic than that in the photosystem I RC.


Assuntos
Helicobacter/metabolismo , Proteínas Ferro-Enxofre/química , Complexo de Proteínas do Centro de Reação Fotossintética/química , Sequência de Aminoácidos , Sítios de Ligação , Membrana Celular/metabolismo , Dimerização , Espectroscopia de Ressonância de Spin Eletrônica , Transporte de Elétrons , Helicobacter/química , Helicobacter/citologia , Helicobacter/genética , Proteínas Ferro-Enxofre/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Complexo de Proteínas do Centro de Reação Fotossintética/genética , Complexo de Proteínas do Centro de Reação Fotossintética/metabolismo , Homologia de Sequência de Aminoácidos , Temperatura
16.
Free Radic Biol Med ; 36(12): 1481-95, 2004 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15182851

RESUMO

Increase in reactive oxygen species plays an integral part in the inflammatory response, and chronic inflammation increases cancer risk. Selenium-dependent glutathione peroxidase (GPX) is well recognized for its antioxidant, and thus anti-inflammatory, activity. However, due to the multiple antioxidant families present in the gastrointestinal tract, it has been difficult to demonstrate the importance of individual antioxidant enzymes. Using genetically altered mice deficient in individual Gpx genes has provided insight into the physiological functions of these genes. Insufficient GPX activity in the mucosal epithelium can trigger acute and chronic inflammation. The presence of certain microflora, such as Helicobacter species, may affect cancer risk significantly. However, when damaged cells have progressed into a precancerous status, increased GPX activity may become procarcinogenic, presumably due to inhibition of hydroperoxide-mediated apoptosis. This review summarizes the current view of GPX in inflammation and cancer with emphasis on the GI tract.


Assuntos
Glutationa Peroxidase/fisiologia , Neoplasias/imunologia , Selênio/metabolismo , Animais , Antioxidantes/metabolismo , Apoptose , Radicais Livres , Gastroenteropatias/metabolismo , Glutationa Peroxidase/metabolismo , Helicobacter/metabolismo , Humanos , Inflamação , Doenças Inflamatórias Intestinais/metabolismo , Camundongos , Camundongos Knockout , Neoplasias/metabolismo , Espécies Reativas de Oxigênio , Selênio/deficiência , Transdução de Sinais , Fatores de Tempo
17.
Proteomics ; 3(3): 273-8, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12627380

RESUMO

Helicobacter pylori is one of the most prevalent human pathogens in the world and is the aetiological agent of gastritis, peptic ulcer disease and gastric malignancies. In addition H. pylori and other novel members of the genus are capable of successfully colonising the bile-rich niche of the upper intestine and are associated with a diverse range of intestinal pathologies. Surface-enhanced laser desorption/ionisation-time of flight mass spectrometry was used to analyse surface extracts from H. pylori, Helicobacter bilis, Helicobacter pullorum and "Helicobacter sp. flexispira" to characterise cell surface changes following bile stress. The system detected two distinct response patterns to bile stress on the cell surface of Helicobacter spp. in vitro. The first involved the increase under bile stress of peaks at 7.6 and 7.9 kDa for H. billis and H. pullorum, respectively. In contrast both "Helicobacter sp. flexispira" and a clinical isolate of H. pylori had similar response profiles to bile stress. Both strains had at least three low mass peaks decreased under bile stress and a single peak induced by bile stress. The present study has established the use of ProteinChip(R) technology to analyse helicobacter-related proteomics. Specifically this study has established that different patterns are generated in response to bile stress among various pathogenic Helicobacter spp. which may give insights into the ability of these strains to colonise different niches.


Assuntos
Bile/química , Helicobacter/metabolismo , Análise Serial de Proteínas/métodos , Proteínas/análise , Gastrite/microbiologia , Humanos , Concentração de Íons de Hidrogênio , Peso Molecular , Proteínas/química , Proteômica , Especificidade da Espécie
18.
Proc Natl Acad Sci U S A ; 100(3): 1292-7, 2003 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-12552104

RESUMO

Long-term colonization of humans with Helicobacter pylori can cause the development of gastric B cell mucosa-associated lymphoid tissue lymphoma, yet little is known about the sequence of molecular steps that accompany disease progression. We used microarray analysis and laser microdissection to identify gene expression profiles characteristic and predictive of the various histopathological stages in a mouse model of the disease. The initial step in lymphoma development is marked by infiltration of reactive lymphocytes into the stomach and the launching of a mucosal immune response. Our analysis uncovered molecular markers of both of these processes, including genes coding for the immunoglobulins and the small proline-rich protein Sprr 2A. The subsequent step is characterized histologically by the antigen-driven proliferation and aggregation of B cells and the gradual appearance of lymphoepithelial lesions. In tissues of this stage, we observed increased expression of genes previously associated with malignancy, including the laminin receptor-1 and the multidrug-resistance channel MDR-1. Finally, we found that the transition to destructive lymphoepithelial lesions and malignant lymphoma is marked by an increase in transcription of a single gene encoding calgranulin AMrp-8.


Assuntos
Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Tecido Linfoide/microbiologia , Linfoma/microbiologia , Mucosa/microbiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Algoritmos , Animais , Calgranulina A/metabolismo , Regulação para Baixo , Helicobacter/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , Regulação para Cima
19.
J Clin Microbiol ; 40(6): 1908-12, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12037042

RESUMO

Helicobacter cinaedi may cause proctocolitis or bacteremia in homosexual men infected with human immunodeficiency virus or occasionally in other immunocompromised hosts. There are scattered reports of H. cinaedi isolated from a variety of animal hosts, but to date only hamsters have been found to be a common natural reservoir. Microaerophillic cultures of feces from 5 of 16 asymptomatic rhesus monkeys (Macaca mulatta) (31%) were positive for a curved gram-negative rod. A polyphasic taxonomic approach was used to identify the organism as H. cinaedi. These results show that H. cinaedi frequently colonizes asymptomatic captive rhesus monkeys, which may serve as another potential reservoir for human infection.


Assuntos
Animais de Laboratório , Infecções por Helicobacter/veterinária , Helicobacter/classificação , Helicobacter/isolamento & purificação , Macaca mulatta , Doenças dos Macacos/microbiologia , Animais , Técnicas de Tipagem Bacteriana , Meios de Cultura , DNA Ribossômico/análise , DNA Ribossômico/genética , Fezes/microbiologia , Feminino , Genótipo , Helicobacter/genética , Helicobacter/metabolismo , Helicobacter/ultraestrutura , Infecções por Helicobacter/microbiologia , Masculino , Dados de Sequência Molecular , Filogenia , RNA Ribossômico 16S/genética , RNA Ribossômico 23S/genética , Análise de Sequência de DNA
20.
DNA Seq ; 13(6): 321-31, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12652903

RESUMO

The urease gene cluster from Helicobacter bizzozeronii was cloned and sequenced. A genomic library was constructed in a lambda-ZAPII vector using TSP5091-digested H. bizzozeronii chromosomal DNA. Four overlapping recombinant bacteriophages carrying the H. bizzozeronii urease genes were identified by using a fragment of H. bizzozeronii ureB as a probe. Sequence analysis of two clones (pHB1 and pHB3) revealed seven open reading frames encoding proteins with predicted masses of 26.5, 60.3, 21.7, 19.5, 28.6, 21.7 and 29.6 kDa representing the structural genes, Urease A and B and its accessory genes, urease I, E, F, G and H, respectively. In addition, three open reading frames upstream of the ureA gene encoding a putative tRNA transferase, a putative Glucose inhibited division protein B (GidB) and a protein with unknown function were also identified. A clone (pHB5) containing a complete urease gene cluster was constructed. The homologue analysis revealed that UreA polypeptide exhibited 64-90% identity to that of Helicobacter heilmanii, Helicobacter felis, Helicobacter pylori, Helicobacter mustelae and Helicobacter hepaticus. UreB polypeptides exhibited 76.8-96% identity to that of H. heilmanii, H. felis, H. pylori, H. mustelae and H. hepaticus. The UreI, E, F, G and H also showed 44-86% identity to that of H. pylori. Among these accessory genes, UreE had a lowest percentage identity to that of H. pylori.


Assuntos
Helicobacter/genética , Família Multigênica , Urease/genética , Sequência de Aminoácidos , Sequência de Bases , Western Blotting , Clonagem Molecular , Helicobacter/metabolismo , Dados de Sequência Molecular , Biblioteca de Peptídeos , Urease/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA