Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Infect Immun ; 88(6)2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32205402

RESUMO

Chronic Helicobacter pylori colonization in animal models often leads to downregulation of the type IV secretion system (T4SS), typically by recombination in cagY, which is an essential T4SS gene. However, 17 other cag pathogenicity island (cagPAI) genes, as well as some non-cagPAI genes, are also essential for T4SS function. To get a more complete picture of how H. pylori regulates the T4SS during animal colonization, we examined cagY in 534 mouse-passaged isolates that lost T4SS function, defined as a normalized interleukin-8 (IL-8) value of <0.3 relative to the input H. pylori strain PMSS1. In order to analyze the genetic changes in the strains with unchanged cagY, we sequenced the entire pathogenicity island of 60 such isolates using single-molecule, real-time (SMRT) sequencing technology (PacBio, Menlo Park, CA), and we compared the results to the PMSS1 wild type (WT). Of the 534 strains, 271 (51%) showed evidence of recombination in cagY, but we also found indels or nonsynonymous changes in 13 other essential cagPAI genes implicated in H. pylori T4SS function, most commonly cag5, cag10, and cagA While cagY recombination is the most common mechanism by which H. pylori downregulates T4SS function during murine infection, loss of function is also associated with changes in other essential cagPAI genes.


Assuntos
Genes Bacterianos , Ilhas Genômicas , Infecções por Helicobacter/microbiologia , Helicobacter pylori/fisiologia , Sistemas de Secreção Tipo IV/genética , Animais , Proteínas de Bactérias/genética , Mapeamento Cromossômico , Camundongos , Recombinação Genética
2.
Infect Immun ; 85(6)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28396320

RESUMO

Most Helicobacter pylori strains express the BabA adhesin, which binds to ABO/Leb blood group antigens on gastric mucin and epithelial cells and is found more commonly in strains that cause peptic ulcers or gastric cancer, rather than asymptomatic infection. We and others have previously reported that in mice, gerbils, and rhesus macaques, expression of babA is lost, either by phase variation or by gene conversion, in which the babB paralog recombines into the babA locus. The functional significance of loss of babA expression is unknown. Here we report that in rhesus monkeys, there is independent selective pressure for loss of babA and for overexpression of BabB, which confers a fitness advantage. Surprisingly, loss of babA by phase variation or gene conversion is not dependent on the capacity of BabA protein to bind Leb, which suggests that it may have other, unrecognized functions. These findings have implications for the role of outer membrane protein diversity in persistent H. pylori infection.


Assuntos
Adesinas Bacterianas/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/patogenicidade , Adesinas Bacterianas/genética , Animais , Aderência Bacteriana , Proteínas da Membrana Bacteriana Externa/genética , Feminino , Aptidão Genética , Genótipo , Helicobacter pylori/metabolismo , Macaca mulatta , Masculino , Mutação , Análise de Sequência de DNA , Estômago/microbiologia , Estômago/patologia
3.
Gastroenterology ; 151(6): 1164-1175.e3, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27569724

RESUMO

BACKGROUND & AIMS: Peptic ulcer disease and gastric cancer are caused most often by Helicobacter pylori strains that harbor the cag pathogenicity island, which encodes a type IV secretion system (T4SS) that injects the CagA oncoprotein into host cells. cagY is an essential gene in the T4SS and has an unusual DNA repeat structure that predicts in-frame insertions and deletions. These cagY recombination events typically lead to a reduction in T4SS function in mouse and primate models. We examined the role of the immune response in cagY-dependent modulation of T4SS function. METHODS: H pylori T4SS function was assessed by measuring CagA translocation and the capacity to induce interleukin (IL)8 in gastric epithelial cells. cagY recombination was determined by changes in polymerase chain reaction restriction fragment-length polymorphisms. T4SS function and cagY in H pylori from C57BL/6 mice were compared with strains recovered from Rag1-/- mice, T- and B-cell-deficient mice, mice with deletion of the interferon gamma receptor (IFNGR) or IL10, and Rag1-/- mice that received adoptive transfer of control or Ifng-/- CD4+ T cells. To assess relevance to human beings, T4SS function and cagY recombination were assessed in strains obtained sequentially from a patient after 7.4 years of infection. RESULTS: H pylori infection of T-cell-deficient and Ifngr1-/- mice, and transfer of CD4+ T cells to Rag1-/- mice, showed that cagY-mediated loss of T4SS function requires a T-helper 1-mediated immune response. Loss of T4SS function and cagY recombination were more pronounced in Il10-/- mice, and in control mice infected with H pylori that expressed a more inflammatory form of cagY. Complementation analysis of H pylori strains isolated from a patient over time showed changes in T4SS function that were dependent on recombination in cagY. CONCLUSIONS: Analysis of H pylori strains from mice and from a chronically infected patient showed that CagY functions as an immune-sensitive regulator of T4SS function. We propose that this is a bacterial adaptation to maximize persistent infection and transmission to a new host under conditions of a robust inflammatory response.


Assuntos
Proteínas de Bactérias/genética , Células Epiteliais/metabolismo , Infecções por Helicobacter/genética , Infecções por Helicobacter/imunologia , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Sistemas de Secreção Tipo IV/genética , Animais , Antígenos de Bactérias/genética , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Doença Crônica , Feminino , Mucosa Gástrica/citologia , Gastrite/imunologia , Gastrite/microbiologia , Infecções por Helicobacter/sangue , Proteínas de Homeodomínio/genética , Humanos , Interferon gama/metabolismo , Interleucina-10/deficiência , Interleucina-10/genética , Interleucina-8/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Recombinação Genética , Transdução de Sinais , Linfócitos T Auxiliares-Indutores , Fatores de Tempo , Translocação Genética , Receptor de Interferon gama
4.
PLoS Pathog ; 9(2): e1003189, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23468628

RESUMO

Helicobacter pylori causes clinical disease primarily in those individuals infected with a strain that carries the cytotoxin associated gene pathogenicity island (cagPAI). The cagPAI encodes a type IV secretion system (T4SS) that injects the CagA oncoprotein into epithelial cells and is required for induction of the pro-inflammatory cytokine, interleukin-8 (IL-8). CagY is an essential component of the H. pylori T4SS that has an unusual sequence structure, in which an extraordinary number of direct DNA repeats is predicted to cause rearrangements that invariably yield in-frame insertions or deletions. Here we demonstrate in murine and non-human primate models that immune-driven host selection of rearrangements in CagY is sufficient to cause gain or loss of function in the H. pylori T4SS. We propose that CagY functions as a sort of molecular switch or perhaps a rheostat that alters the function of the T4SS and "tunes" the host inflammatory response so as to maximize persistent infection.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Sistemas de Secreção Bacterianos/fisiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/metabolismo , Animais , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , DNA Bacteriano , Feminino , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/metabolismo , Helicobacter pylori/imunologia , Helicobacter pylori/ultraestrutura , Interações Hospedeiro-Patógeno , Interleucina-8/metabolismo , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Varredura , Recombinação Genética , Organismos Livres de Patógenos Específicos , Fatores de Virulência
5.
bioRxiv ; 2023 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-37461695

RESUMO

Most cases of gastric cancer are caused by chronic Helicobacter pylori infection, but the lack of early onco-diagnostics and a high risk for antibiotic resistance hampers early intervention through eradication of H. pylori infection by antibiotics. We reported on a protective mechanism where H. pylori gastric mucosal attachment can be reduced by natural antibodies that block the binding of its attachment protein BabA. Here we show that challenge infection with H. pylori induced response of such blocking antibodies in both human volunteers and in rhesus macaques, that mucosal vaccination with BabA protein antigen induced blocking antibodies in rhesus macaques, and that vaccination in a mouse model induced blocking antibodies that reduced gastric mucosal inflammation, preserved the gastric juice acidity, and fully protected the mice from gastric cancer caused by H. pylori.

6.
Infect Immun ; 80(2): 594-601, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22124657

RESUMO

The best-studied Helicobacter pylori virulence factor associated with development of peptic ulcer disease or gastric cancer (GC) rather than asymptomatic nonatrophic gastritis (NAG) is the cag pathogenicity island (cagPAI), which encodes a type IV secretion system (T4SS) that injects the CagA oncoprotein into host epithelial cells. Here we used real-time reverse transcription-PCR (RT-PCR) to measure the in vivo expression of genes on the cagPAI and of other virulence genes in patients with NAG, duodenal ulcer (DU), or GC. In vivo expression of H. pylori virulence genes was greater overall in gastric biopsy specimens of patients with GC than in those of patients with NAG or DU. However, since in vitro expression of cagA was not greater in H. pylori strains from patients with GC than in those from patients with NAG or DU, increased expression in GC in vivo is likely a result of environmental conditions in the gastric mucosa, though it may in turn cause more severe pathology. Increased expression of virulence genes in GC may represent a stress response to elevated pH or other environmental conditions in the stomach of patients with GC, which may be less hospitable to H. pylori colonization than the acidic environment in patients with NAG or DU.


Assuntos
Úlcera Duodenal/microbiologia , Gastrite/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Helicobacter pylori/patogenicidade , Neoplasias Gástricas/microbiologia , Adulto , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Mucosa Gástrica/microbiologia , Regulação Bacteriana da Expressão Gênica , Humanos , Virulência/genética
7.
Gut Microbes ; 14(1): 2044721, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35289715

RESUMO

Helicobacter pylori is the major risk factor for gastric cancer. H. pylori harboring the type IV secretion system (T4SS) and its effector CagA encoded on the cag pathogenicity Island (cagPAI) increases the risk. H. pylori PMSS1 has a multi-cagA genotype, modulating cagA copy number dynamically from zero to four copies. To examine the effect of the immune response on cagA copy number change, we utilized a mouse model with different immune status. PMSS1 recovered from Rag1-/- mice, lacking functional T or B cells, retained more cagA copies. PMSS1 recovered from Il10-/- mice, showing intense inflammation, had fewer cagA copies compared to those recovered from wild-type mice. Moreover, cagA copy number of PMSS1 recovered from wild-type and Il10-/- mice was positively correlated with the capacity to induce IL-8 secretion at four weeks of infection. Since recombination in cagY influences T4SS function, including CagA translocation and IL-8 induction, we constructed a multiple linear regression model to predict H. pylori-induced IL-8 expression based on cagA copy number and cagY recombination status; H. pylori induces more IL-8 secretion when the strain has more cagA copies and intact cagY. This study shows that H. pylori PMSS1 in mice with less intense immune response possess higher cagA copy number than those infected in mice with more intense immune response and thus the multi-cagA genotype, along with cagY recombination, functions as an immune-sensitive regulator of H. pylori virulence.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Microbioma Gastrointestinal , Infecções por Helicobacter , Helicobacter pylori , Animais , Proteínas de Bactérias/metabolismo , Variações do Número de Cópias de DNA , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Helicobacter pylori/patogenicidade , Imunidade , Interleucina-10/genética , Interleucina-8/metabolismo , Camundongos , Virulência
8.
Infect Immun ; 78(4): 1593-600, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20123715

RESUMO

The Helicobacter pylori babA gene encodes an outer membrane protein that mediates binding to fucosylated ABH antigens of the ABO blood group. We recently demonstrated that BabA expression is lost during experimental infection of rhesus macaques with H. pylori J166. We sought to test the generality of this observation by comparison of different H. pylori strains and different animal hosts. Challenge of macaques with H. pylori J99 yielded output strains that lost BabA expression, either by selection and then expansion of a subpopulation of J99 that had a single-base-pair mutation that encoded a stop codon or by gene conversion of babA with a duplicate copy of babB, a paralog of unknown function. Challenge of mice with H. pylori J166, which unlike J99, has 5' CT repeats in babA, resulted in loss of BabA expression due to phase variation. In the gerbil, Leb binding was lost by replacement of the babA gene that encoded Leb binding with a nonbinding allele that differed at six amino acid residues. Complementation experiments confirmed that change in these six amino acids of BabA was sufficient to eliminate binding to Leb and to gastric tissue. These results demonstrate that BabA expression in vivo is highly dynamic, and the findings implicate specific amino acid residues as critical for binding to fucosylated ABH antigens. We hypothesize that modification of BabA expression during H. pylori infection is a mechanism to adapt to changing conditions of inflammation and glycan expression at the epithelial surface.


Assuntos
Adesinas Bacterianas/biossíntese , Aderência Bacteriana , Infecções por Helicobacter/microbiologia , Helicobacter pylori/patogenicidade , Adaptação Biológica , Adesinas Bacterianas/genética , Animais , DNA Bacteriano/química , DNA Bacteriano/genética , Feminino , Mucosa Gástrica/microbiologia , Técnicas de Inativação de Genes , Teste de Complementação Genética , Gerbillinae , Helicobacter pylori/genética , Helicobacter pylori/isolamento & purificação , Humanos , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutação , Análise de Sequência de DNA
9.
mBio ; 11(6)2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33443133

RESUMO

The Helicobacter pylori type IV secretion system (T4SS) encoded on the cag pathogenicity island (cagPAI) secretes the CagA oncoprotein and other effectors into the gastric epithelium. During murine infection, T4SS function is lost in an immune-dependent manner, typically as a result of in-frame recombination in the middle repeat region of cagY, though single nucleotide polymorphisms (SNPs) in cagY or in other essential genes may also occur. Loss of T4SS function also occurs in gerbils, nonhuman primates, and humans, suggesting that it is biologically relevant and not simply an artifact of the murine model. Here, we sought to identify physiologically relevant conditions under which T4SS function is maintained in the murine model. We found that loss of H. pylori T4SS function in mice was blunted by systemic Salmonella coinfection and completely eliminated by dietary iron restriction. Both have epidemiologic parallels in humans, since H. pylori strains from individuals in developing countries, where iron deficiency and systemic infections are common, are also more often cagPAI+ than strains from developed countries. These results have implications for our fundamental understanding of the cagPAI and also provide experimental tools that permit the study of T4SS function in the murine model.IMPORTANCE The type IV secretion system (T4SS) is the major Helicobacter pylori virulence factor, though its function is lost during murine infection. Loss of function also occurs in gerbils and in humans, suggesting that it is biologically relevant, but the conditions under which T4SS regulation occurs are unknown. Here, we found that systemic coinfection with Salmonella and iron deprivation each promote retention of T4SS function. These results improve our understanding of the cag pathogenicity island (cagPAI) and provide experimental tools that permit the study of T4SS function in the murine model.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Ilhas Genômicas , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Sistemas de Secreção Tipo IV/genética , Animais , Coinfecção/microbiologia , Feminino , Mucosa Gástrica , Helicobacter pylori/metabolismo , Helicobacter pylori/patogenicidade , Ferro/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Salmonelose Animal/sangue , Salmonelose Animal/microbiologia , Sistemas de Secreção Tipo IV/metabolismo , Fatores de Virulência
10.
Mol Microbiol ; 69(4): 994-1007, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18573180

RESUMO

Helicobacter pylori colonization of the human stomach is characterized by profound disease-causing inflammation. Bacterial proteins that detoxify reactive oxygen species or recognize damaged DNA adducts promote infection, suggesting that H. pylori requires DNA damage repair for successful in vivo colonization. The molecular mechanisms of repair remain unknown. We identified homologues of the AddAB class of helicase-nuclease enzymes, related to the Escherichia coli RecBCD enzyme, which, with RecA, is required for repair of DNA breaks and homologous recombination. H. pylori mutants lacking addA or addB genes lack detectable ATP-dependent nuclease activity, and the cloned H. pylori addAB genes restore both nuclease and helicase activities to an E. coli recBCD deletion mutant. H. pylori addAB and recA mutants have a reduced capacity for stomach colonization. These mutants are sensitive to DNA damaging agents and have reduced frequencies of apparent gene conversion between homologous genes encoding outer membrane proteins. Our results reveal requirements for double-strand break repair and recombination during both acute and chronic phases of H. pylori stomach infection.


Assuntos
Proteínas de Bactérias/fisiologia , DNA Helicases/fisiologia , Exodesoxirribonucleases/fisiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/fisiologia , Recombinases Rec A/fisiologia , Estômago/microbiologia , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Quebras de DNA de Cadeia Dupla , DNA Helicases/genética , Reparo do DNA , Escherichia coli/enzimologia , Escherichia coli/genética , Exodesoxirribonucleases/genética , Feminino , Helicobacter pylori/enzimologia , Helicobacter pylori/genética , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Recombinases Rec A/genética , Recombinação Genética
11.
mBio ; 9(3)2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29764950

RESUMO

Strains of Helicobacter pylori that cause ulcer or gastric cancer typically express a type IV secretion system (T4SS) encoded by the cag pathogenicity island (cagPAI). CagY is an ortholog of VirB10 that, unlike other VirB10 orthologs, has a large middle repeat region (MRR) with extensive repetitive sequence motifs, which undergo CD4+ T cell-dependent recombination during infection of mice. Recombination in the CagY MRR reduces T4SS function, diminishes the host inflammatory response, and enables the bacteria to colonize at a higher density. Since CagY is known to bind human α5ß1 integrin, we tested the hypothesis that recombination in the CagY MRR regulates T4SS function by modulating binding to α5ß1 integrin. Using a cell-free microfluidic assay, we found that H. pylori binding to α5ß1 integrin under shear flow is dependent on the CagY MRR, but independent of the presence of the T4SS pili, which are only formed when H. pylori is in contact with host cells. Similarly, expression of CagY in the absence of other T4SS genes was necessary and sufficient for whole bacterial cell binding to α5ß1 integrin. Bacteria with variant cagY alleles that reduced T4SS function showed comparable reduction in binding to α5ß1 integrin, although CagY was still expressed on the bacterial surface. We speculate that cagY-dependent modulation of H. pylori T4SS function is mediated by alterations in binding to α5ß1 integrin, which in turn regulates the host inflammatory response so as to maximize persistent infection.IMPORTANCE Infection with H. pylori can cause peptic ulcers and is the most important risk factor for gastric cancer, the third most common cause of cancer death worldwide. The major H. pylori virulence factor that determines whether infection causes disease or asymptomatic colonization is the type IV secretion system (T4SS), a sort of molecular syringe that injects bacterial products into gastric epithelial cells and alters host cell physiology. We previously showed that recombination in CagY, an essential T4SS component, modulates the function of the T4SS. Here we found that these recombination events produce parallel changes in specific binding to α5ß1 integrin, a host cell receptor that is essential for T4SS-dependent translocation of bacterial effectors. We propose that CagY-dependent binding to α5ß1 integrin acts like a molecular rheostat that alters T4SS function and modulates the host immune response to promote persistent infection.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Integrina alfa5/metabolismo , Integrina beta1/metabolismo , Sistemas de Secreção Tipo IV/metabolismo , Motivos de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Ilhas Genômicas , Infecções por Helicobacter/genética , Infecções por Helicobacter/microbiologia , Helicobacter pylori/química , Helicobacter pylori/genética , Interações Hospedeiro-Patógeno , Humanos , Integrina alfa5/genética , Integrina beta1/genética , Ligação Proteica , Sistemas de Secreção Tipo IV/genética
12.
PLoS One ; 12(8): e0183324, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28813514

RESUMO

Epidemiologic studies have reported an inverse relationship between childhood Helicobacter pylori infection and development of allergic asthma. Because lung epithelium plays an important role in allergic asthma pathogenesis, we hypothesized that H. pylori may directly influence airway epithelial cell innate immune function, particularly in early childhood. To test our hypothesis, we established an in vitro H. pylori infection model using primary tracheobronchial epithelial cell cultures derived from infant, juvenile and adult rhesus monkeys. Airway epithelial cell cultures were infected with wild-type or cag pathogenicity island mutant H. pylori strains, followed by evaluation of IL-8 and IL-6 protein synthesis. We found that H. pylori primarily increased IL-8 synthesis in a MOI and age-dependent fashion, with a greater than 4-fold induction in infant versus adult cultures. H. pylori-induced IL-8 synthesis in infant and juvenile cultures was significantly reduced by cag pathogenicity island mutants, indicating a requirement for the type IV secretion system. Although peptidoglycan recognition of nucleotide binding oligomerization domain-containing protein 1 (NOD1) and NF-kappaB have been implicated as key cytokine signaling molecules for H. pylori infection in gastric epithelium, NOD1 (ML130) or NF-kappaB (JSH-23) inhibitors minimally affected IL-8 synthesis in airway epithelial cell cultures following H. pylori infection. In contrast, inhibition of the p38 MAP kinase pathway (SB203580) resulted in almost complete suppression of H. pylori-induced IL-8 synthesis. Collectively, these results indicate that H. pylori can preferentially elicit IL-8 synthesis in a model of pediatric airway epithelium using the type IV secretion system via p38 MAP kinase.


Assuntos
Helicobacter pylori/fisiologia , Interleucina-8/metabolismo , Mucosa Respiratória/imunologia , Mucosa Respiratória/microbiologia , Sistemas de Secreção Tipo IV/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Linhagem Celular , Infecções por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Humanos , Técnicas In Vitro , Interleucina-6/metabolismo , Primatas , Mucosa Respiratória/enzimologia , Transdução de Sinais/fisiologia
13.
Sci Rep ; 7: 46499, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28418004

RESUMO

Expression of the Helicobacter pylori blood group antigen binding adhesin A (BabA) is more common in strains isolated from patients with peptic ulcer disease or gastric cancer, rather than asymptomatic colonization. Here we used mouse models to examine host determinants that affect H. pylori BabA expression. BabA expression was lost by phase variation as frequently in WT mice as in RAG2-/- mice that do not have functional B or T cells, and in MyD88-/-, TLR2-/- and TLR4-/- mice that are defective in toll like receptor signaling. The presence of other bacteria had no effect on BabA expression as shown by infection of germ free mice. Moreover, loss of BabA expression was not dependent on Leb expression or the capacity of BabA to bind Leb. Surprisingly, gender was the host determinant most associated with loss of BabA expression, which was maintained to a greater extent in male mice and was associated with greater bacterial load. These results suggest the possibility that loss of BabA expression is not driven by adaptive immunity or toll-like receptor signaling, and that BabA may have other, unrecognized functions in addition to serving as an adhesin that binds Leb.


Assuntos
Adesinas Bacterianas/biossíntese , Regulação Bacteriana da Expressão Gênica , Infecções por Helicobacter/metabolismo , Helicobacter pylori/fisiologia , Interações Hospedeiro-Patógeno , Adesinas Bacterianas/genética , Animais , Modelos Animais de Doenças , Feminino , Infecções por Helicobacter/microbiologia , Humanos , Masculino , Camundongos , Camundongos Knockout
14.
mBio ; 8(1)2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28223462

RESUMO

Many bacterial genomes are highly variable but nonetheless are typically published as a single assembled genome. Experiments tracking bacterial genome evolution have not looked at the variation present at a given point in time. Here, we analyzed the mouse-passaged Helicobacter pylori strain SS1 and its parent PMSS1 to assess intra- and intergenomic variability. Using high sequence coverage depth and experimental validation, we detected extensive genome plasticity within these H. pylori isolates, including movement of the transposable element IS607, large and small inversions, multiple single nucleotide polymorphisms, and variation in cagA copy number. The cagA gene was found as 1 to 4 tandem copies located off the cag island in both SS1 and PMSS1; this copy number variation correlated with protein expression. To gain insight into the changes that occurred during mouse adaptation, we also compared SS1 and PMSS1 and observed 46 differences that were distinct from the within-genome variation. The most substantial was an insertion in cagY, which encodes a protein required for a type IV secretion system function. We detected modifications in genes coding for two proteins known to affect mouse colonization, the HpaA neuraminyllactose-binding protein and the FutB α-1,3 lipopolysaccharide (LPS) fucosyltransferase, as well as genes predicted to modulate diverse properties. In sum, our work suggests that data from consensus genome assemblies from single colonies may be misleading by failing to represent the variability present. Furthermore, we show that high-depth genomic sequencing data of a population can be analyzed to gain insight into the normal variation within bacterial strains.IMPORTANCE Although it is well known that many bacterial genomes are highly variable, it is nonetheless traditional to refer to, analyze, and publish "the genome" of a bacterial strain. Variability is usually reduced ("only sequence from a single colony"), ignored ("just publish the consensus"), or placed in the "too-hard" basket ("analysis of raw read data is more robust"). Now that whole-genome sequences are regularly used to assess virulence and track outbreaks, a better understanding of the baseline genomic variation present within single strains is needed. Here, we describe the variability seen in typical working stocks and colonies of pathogen Helicobacter pylori model strains SS1 and PMSS1 as revealed by use of high-coverage mate pair next-generation sequencing (NGS) and confirmed by traditional laboratory techniques. This work demonstrates that reliance on a consensus assembly as "the genome" of a bacterial strain may be misleading.


Assuntos
Variação Genética , Genoma Bacteriano , Helicobacter pylori/genética , Animais , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Mutação
15.
J Am Vet Med Assoc ; 228(5): 751-5, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16506942

RESUMO

OBJECTIVE: To determine molecular characteristics, antimicrobial susceptibility, and toxigenicity of Clostridium difficile isolates from horses in an intensive care unit and evaluate associations among severity of clinical disease with specific strains of C difficile. DESIGN: Prospective study. ANIMALS: 130 horses. PROCEDURES: Feces were collected from horses admitted for acute gastrointestinal tract disease with loose feces and submitted for microbial culture and immunoassay for toxin production. Polymerase chain reaction assays were performed on isolates for toxins A and B genes and strain identification. RESULTS: Isolates were grouped into 3 strains (A, B, and C) on the basis of molecular banding patterns. Toxins A and B gene sequences were detected in 93%, 95%, and 73% of isolates of strains A, B, and C, respectively. Results of fecal immunoassays for toxin A were positive in 40%, 63%, and 16% of horses with strains A, B, and C, respectively. Isolates in strain B were resistant to metronidazole. Horses infected with strain B were 10 times as likely to have been treated with metronidazole prior to the onset of diarrhea as horses infected with other strains. Duration from onset of diarrhea to discharge (among survivors) was longer, systemic inflammatory response syndromes were more pronounced, and mortality rate was higher in horses infected with strain B than those infected with strains A and C combined. CONCLUSIONS AND CLINICAL RELEVANCE: Horses may be infected with a number of heterogeneous isolates of C difficile. Results indicated that toxigenicity and antimicrobial susceptibility of isolates vary and that metronidazole-resistant strains may be associated with severe disease.


Assuntos
Antibacterianos/uso terapêutico , Toxinas Bacterianas/análise , Clostridioides difficile , Enterocolite Pseudomembranosa/microbiologia , Doenças dos Cavalos/microbiologia , Animais , Antibacterianos/farmacologia , Toxinas Bacterianas/química , Sequência de Bases , Clostridioides difficile/classificação , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/isolamento & purificação , Farmacorresistência Bacteriana , Enterocolite Pseudomembranosa/tratamento farmacológico , Fezes/microbiologia , Feminino , Doenças dos Cavalos/tratamento farmacológico , Cavalos , Hospitais Veterinários , Masculino , Metronidazol/farmacologia , Metronidazol/uso terapêutico , Testes de Sensibilidade Microbiana/veterinária , Filogenia , Reação em Cadeia da Polimerase/métodos , Reação em Cadeia da Polimerase/veterinária , Estudos Prospectivos , Índice de Gravidade de Doença
16.
FEMS Microbiol Lett ; 363(24)2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27940463

RESUMO

Helicobacter pylori commonly infects the epithelial layer of the human stomach and in some individuals causes peptic ulcers, gastric adenocarcinoma or gastric lymphoma. Helicobacter pylori is a genetically diverse species, and the most important bacterial virulence factor that increases the risk of developing disease, versus asymptomatic colonization, is the cytotoxin associated gene pathogenicity island (cagPAI). Socially housed rhesus macaques are often naturally infected with H. pylori similar to that which colonizes humans, but little is known about the cagPAI. Here we show that H. pylori strains isolated from naturally infected rhesus macaques have a cagPAI very similar to that found in human clinical isolates, and like human isolates, it encodes a functional type IV secretion system. These results provide further support for the relevance of rhesus macaques as a valid experimental model for H. pylori infection in humans.


Assuntos
Ilhas Genômicas , Infecções por Helicobacter/veterinária , Helicobacter pylori/genética , Macaca mulatta , Doenças dos Primatas/microbiologia , Animais , Genes Bacterianos , Infecções por Helicobacter/microbiologia , Helicobacter pylori/isolamento & purificação , Homologia de Sequência , Sistemas de Secreção Tipo IV/genética
17.
J Am Vet Med Assoc ; 220(1): 67-73, 2002 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12680451

RESUMO

OBJECTIVE: To determine molecular characteristics of Clostridium difficile isolates from foals with diarrhea and identify clinical abnormalities in affected foals. DESIGN: Retrospective study. ANIMALS: 28 foals with C difficile-associated diarrhea. PROCEDURE: Toxigenicity, molecular fingerprinting, and antibiotic susceptibility patterns were determined. Information on signalment, clinical findings, results of clinicopathologic testing, whether antimicrobials had been administered prior to development of diarrhea, and outcome was obtained from the medical records. RESULTS: Twenty-three (82%) foals survived. Toxin A and B gene sequences were detected in isolates from 24 of 27 foals, whereas the toxin B gene alone was detected in the isolate from 1 foal. Results of an ELISA for toxin A were positive for fecal samples from only 8 of 20 (40%) foals. Ten of 23 (43%) isolates were resistant to metronidazole. Molecular fingerprinting revealed marked heterogeneity among isolates, except for the metronidazole-resistant isolates. Sixteen foals had tachypnea. Hematologic abnormalities were indicative of inflammation. Common serum biochemical abnormalities included metabolic acidosis, hyponatremia, hypocalcemia, azotemia, hypoproteinemia, hyperglycemia, and high enzyme activities. Passive transfer of maternal antibodies was adequate in all 12 foals evaluated. CONCLUSIONS AND CLINICAL RELEVANCE: Results suggest that a large percentage of C difficile isolates from foals with diarrhea will have the toxin A and B gene sequences. Because of the possibility that isolates will be resistant to metronidazole, susceptibility testing is warranted. Clostridium difficile isolates from foals may have a substantial amount of molecular heterogeneity. Clinical and hematologic findings in affected foals are similar to those for foals with diarrhea caused by other pathogens.


Assuntos
Proteínas de Bactérias , Clostridioides difficile , Infecções por Clostridium/veterinária , Diarreia/veterinária , Doenças dos Cavalos/microbiologia , Animais , Antibacterianos/uso terapêutico , Toxinas Bacterianas/análise , Toxinas Bacterianas/biossíntese , Toxinas Bacterianas/genética , Clostridioides difficile/classificação , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/fisiologia , Infecções por Clostridium/microbiologia , Diarreia/microbiologia , Farmacorresistência Bacteriana , Enterotoxinas/análise , Enterotoxinas/genética , Ensaio de Imunoadsorção Enzimática/veterinária , Fezes/química , Feminino , Cavalos , Masculino , Metronidazol/uso terapêutico , Testes de Sensibilidade Microbiana/veterinária , Reação em Cadeia da Polimerase/veterinária , Estudos Retrospectivos
18.
Nat Commun ; 5: 4165, 2014 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-24924186

RESUMO

The evolution rate and genetic changes that occur during chronic infection with Helicobacter pylori have been analysed, but little is known about the genomic changes during the initial, acute bacterial infection phase. Here we analyse the rate and pattern of genome evolution in H. pylori from the genomes of two input strains isolated from human volunteers with asymptomatic infection, and the genomes of two output strains collected 20 and 44 days after re-infection. Similarly, we analyse genome evolution in bacteria from the genome sequences of input and output strains sequentially taken after experimental infection of a rhesus macaque. The estimated mutation rate reveals a mutation burst during the acute infection phase that is over 10 times faster than the mutation rate during chronic infection, and orders of magnitude faster than mutation rates in any other bacteria. The elevated frequency of mutations in outer membrane protein genes suggests that the mutation burst facilitates rapid host adaptation of the bacteria.


Assuntos
Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Mutação , Animais , Evolução Molecular , Feminino , Genoma Bacteriano , Helicobacter pylori/fisiologia , Humanos , Macaca mulatta , Dados de Sequência Molecular , Taxa de Mutação
19.
Artigo em Inglês | MEDLINE | ID: mdl-22919637

RESUMO

The Helicobacter pyloricag pathogenicity island (cag PAI) encodes a type IV secretion system that is more commonly found in strains isolated from patients with gastroduodenal disease than from those with asymptomatic gastritis. Genome-wide organization of the transcriptional units in H. pylori strain 26695 was recently established using RNA sequence analysis (Sharma et al., 2010). Here we used quantitative reverse-transcription polymerase chain reaction of open reading frames and intergenic regions to identify putative cag PAI operons in H. pylori; these operons were analyzed further by transcript profiling after deletion of selected promoter regions. Additionally, we used a promoter-trap system to identify functional cag PAI promoters. The results demonstrated that expression of genes on the H. pyloricag PAI varies by nearly five orders of magnitude and that the organization of cag PAI genes into transcriptional units is conserved among several H. pylori strains, including, 26695, J99, G27, and J166. We found evidence for 20 transcripts within the cag PAI, many of which likely overlap. Our data suggests that there are at least 11 operons: cag1-4, cag3-4, cag10-9, cag8-7, cag6-5, cag11-12, cag16-17, cag19-18, cag21-20, cag23-22, and cag25-24, as well as five monocistronic genes (cag4, cag13, cag14, cag15, and cag26). Additionally, the location of four of our functionally identified promoters suggests they are directing expression of, in one case, a truncated version of cag26 and in the other three, transcripts that are antisense to cag7, cag17, and cag23. We verified expression of two of these antisense transcripts, those antisense to cag17 and cag23, by reverse-transcription polymerase chain reaction. Taken together, our results suggest that the cag PAI transcriptional profile is generally conserved among H. pylori strains, 26695, J99, G27, and J166, and is likely complex.


Assuntos
Regulação Bacteriana da Expressão Gênica , Ilhas Genômicas , Helicobacter pylori/genética , Transcrição Gênica , Perfilação da Expressão Gênica , Ordem dos Genes , Óperon , Reação em Cadeia da Polimerase em Tempo Real
20.
PLoS One ; 5(1): e8804, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-20098711

RESUMO

BACKGROUND: Helicobacter pylori, a lifelong and typically asymptomatic infection of the stomach, profoundly alters gastric immune responses, and may benefit the host in protection against other pathogens. We explored the hypothesis that H. pylori contributes to the control of infection with Mycobacterium tuberculosis. METHODOLOGY/PRINCIPAL FINDINGS: We first examined M. tuberculosis-specific IFN-gamma and H. pylori antibody responses in 339 healthy Northern Californians undergoing routine tuberculin skin testing. Of 97 subjects (29%) meeting criteria for latent tuberculosis (TB) infection (LTBI), 45 (46%) were H. pylori seropositive. Subjects with LTBI who were H. pylori-seropositive had 1.5-fold higher TB antigen-induced IFN-gamma responses (p = 0.04, ANOVA), and a more Th-1 like cytokine profile in peripheral blood mononuclear cells, compared to those who were H. pylori seronegative. To explore an association between H. pylori infection and clinical outcome of TB exposure, we evaluated H. pylori seroprevalence in baseline samples from two high risk TB case-contact cohorts, and from cynomolgus macaques experimentally challenged with M. tuberculosis. Compared to 513 household contacts who did not progress to active disease during a median 24 months follow-up, 120 prevalent TB cases were significantly less likely to be H. pylori infected (AOR: 0.55, 95% CI 0.0.36-0.83, p = 0.005), though seroprevalence was not significantly different from non-progressors in 37 incident TB cases (AOR: 1.35 [95% CI 0.63-2.9] p = 0.44). Cynomolgus macaques with natural H. pylori infection were significantly less likely to progress to TB 6 to 8 months after M. tuberculosis challenge (RR: 0.31 [95% CI 0.12-0.80], p = 0.04). CONCLUSIONS/SIGNIFICANCE: H. pylori infection may induce bystander effects that modify the risk of active TB in humans and non-human primates. That immunity to TB may be enhanced by exposure to other microbial agents may have important implications for vaccine development and disease control.


Assuntos
Infecções por Helicobacter/microbiologia , Helicobacter pylori/isolamento & purificação , Tuberculose/complicações , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Animais , Anticorpos Antibacterianos/biossíntese , Estudos de Casos e Controles , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Infecções por Helicobacter/complicações , Infecções por Helicobacter/imunologia , Humanos , Interferon gama/biossíntese , Macaca fascicularis , Masculino , Pessoa de Meia-Idade , Teste Tuberculínico , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa