Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Biol ; 17(5): e3000231, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31048876

RESUMO

Lifelong infection of the gastric mucosa by Helicobacter pylori can lead to peptic ulcers and gastric cancer. However, how the bacteria maintain chronic colonization in the face of constant mucus and epithelial cell turnover in the stomach is unclear. Here, we present a new model of how H. pylori establish and persist in stomach, which involves the colonization of a specialized microenvironment, or microniche, deep in the gastric glands. Using quantitative three-dimensional (3D) confocal microscopy and passive CLARITY technique (PACT), which renders tissues optically transparent, we analyzed intact stomachs from mice infected with a mixture of isogenic, fluorescent H. pylori strains with unprecedented spatial resolution. We discovered that a small number of bacterial founders initially establish colonies deep in the gastric glands and then expand to colonize adjacent glands, forming clonal population islands that persist over time. Gland-associated populations do not intermix with free-swimming bacteria in the surface mucus, and they compete for space and prevent newcomers from establishing in the stomach. Furthermore, bacterial mutants deficient in gland colonization are outcompeted by wild-type (WT) bacteria. Finally, we found that host factors such as the age at infection and T-cell responses control bacterial density within the glands. Collectively, our results demonstrate that microniches in the gastric glands house a persistent H. pylori reservoir, which we propose replenishes the more transient bacterial populations in the superficial mucosa.


Assuntos
Mucosa Gástrica/microbiologia , Helicobacter pylori/crescimento & desenvolvimento , Microscopia Confocal/métodos , Animais , Antibacterianos/farmacologia , Linhagem Celular Tumoral , Contagem de Colônia Microbiana , Feminino , Mucosa Gástrica/efeitos dos fármacos , Infecções por Helicobacter/microbiologia , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/genética , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Especificidade da Espécie , Linfócitos T/efeitos dos fármacos
2.
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
3.
Curr Top Microbiol Immunol ; 421: 21-52, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31123884

RESUMO

Helicobacter pylori chronically infects nearly half the world's population, yet most of those infected remain asymptomatic throughout their lifetime. The outcome of infection-peptic ulcer disease or gastric cancer versus asymptomatic colonization-is a product of host genetics, environmental influences, and differences in bacterial virulence factors. Here, we review the current understanding of the cag pathogenicity island (cagPAI), the vacuolating cytotoxin (VacA), and a large family of outer membrane proteins (OMPs), which are among the best understood H. pylori virulence determinants that contribute to disease. Each of these virulence factors is characterized by allelic and phenotypic diversity that is apparent within and across individuals, as well as over time, and modulates inflammation. From the bacterial perspective, inflammation is probably a necessary evil because it promotes nutrient acquisition, but at the cost of reduction in bacterial load and therefore decreases the chance of transmission to a new host. The general picture that emerges is one of a chronic bacterial infection that is dependent on both inducing and carefully regulating the host inflammatory response. A better understanding of these regulatory mechanisms may have implications for the control of chronic inflammatory diseases that are increasingly common causes of human morbidity and mortality.


Assuntos
Infecções por Helicobacter/imunologia , Infecções por Helicobacter/patologia , Helicobacter pylori/imunologia , Helicobacter pylori/patogenicidade , Estômago/microbiologia , Estômago/patologia , Fatores de Virulência , Proteínas de Bactérias , Infecções por Helicobacter/microbiologia , Humanos , Úlcera Péptica/microbiologia , Úlcera Péptica/patologia , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia
4.
Clin Infect Dis ; 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38843051
5.
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
6.
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
7.
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
8.
J Bacteriol ; 196(2): 337-44, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24187089

RESUMO

Helicobacter pylori uses natural competence and homologous recombination to adapt to the dynamic environment of the stomach mucosa and maintain chronic colonization. Although H. pylori competence is constitutive, its rate of transformation is variable, and little is known about factors that influence it. To examine this, we first determined the transformation efficiency of H. pylori strains under low O2 (5% O2, 7.6% CO2, 7.6% H2) and high O2 (15% O2, 2.9% CO2, 2.9% H2) conditions using DNA containing an antibiotic resistance marker. H. pylori transformation efficiency was 6- to 32-fold greater under high O2 tension, which was robust across different H. pylori strains, genetic loci, and bacterial growth phases. Since changing the O2 concentration for these initial experiments also changed the concentrations of CO2 and H2, transformations were repeated under conditions where O2, CO2, and H2 were each varied individually. The results showed that the increase in transformation efficiency under high O2 was largely due to a decrease in CO2. An increase in pH similar to that caused by low CO2 was also sufficient to increase transformation efficiency. These results have implications for the physiology of H. pylori in the gastric environment, and they provide optimized conditions for the laboratory construction of H. pylori mutants using natural transformation.


Assuntos
Competência de Transformação por DNA/efeitos dos fármacos , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/fisiologia , Transformação Bacteriana , Dióxido de Carbono/metabolismo , Helicobacter pylori/genética , Hidrogênio/metabolismo , Oxigênio/metabolismo
9.
Sci Rep ; 14(1): 9998, 2024 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-38693196

RESUMO

It is estimated that more than half of the world population has been infected with Helicobacter pylori. Most newly acquired H. pylori infections occur in children before 10 years of age. We hypothesized that early life H. pylori infection could influence the composition of the microbiome at mucosal sites distant to the stomach. To test this hypothesis, we utilized the infant rhesus macaque monkey as an animal model of natural H. pylori colonization to determine the impact of infection on the lung and oral microbiome during a window of postnatal development. From a cohort of 4-7 month-old monkeys, gastric biopsy cultures identified 44% of animals infected by H. pylori. 16S ribosomal RNA gene sequencing of lung washes and buccal swabs from animals showed distinct profiles for the lung and oral microbiome, independent of H. pylori infection. In order of relative abundance, the lung microbiome was dominated by the phyla Proteobacteria, Firmicutes, Bacteroidota, Fusobacteriota, Campilobacterota and Actinobacteriota while the oral microbiome was dominated by Proteobacteria, Firmicutes, Bacteroidota, and Fusobacteriota. In comparison to the oral cavity, the lung was composed of more genera and species that significantly differed by H. pylori status, with a total of 6 genera and species that were increased in H. pylori negative infant monkey lungs. Lung, but not plasma IL-8 concentration was also associated with gastric H. pylori load and lung microbial composition. We found the infant rhesus macaque monkey lung harbors a microbiome signature that is distinct from that of the oral cavity during postnatal development. Gastric H. pylori colonization and IL-8 protein were linked to the composition of microbial communities in the lung and oral cavity. Collectively, these findings provide insight into how H. pylori infection might contribute to the gut-lung axis during early childhood and modulate future respiratory health.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Pulmão , Macaca mulatta , Microbiota , Boca , RNA Ribossômico 16S , Animais , Macaca mulatta/microbiologia , Pulmão/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Boca/microbiologia , RNA Ribossômico 16S/genética , Masculino , Modelos Animais de Doenças
10.
J Clin Microbiol ; 51(1): 278-80, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23052320

RESUMO

In a large prospective comparison, the illumigene test detected Clostridium difficile in 98% of toxin-positive and 58% of toxin-negative samples confirmed positive by other methods. The Xpert was uniformly sensitive. Most samples with discrepant results had C. difficile concentrations below the illumigene limit of detection. The significance of low-level C. difficile detection needs investigation.


Assuntos
Carga Bacteriana/métodos , Clostridioides difficile/isolamento & purificação , Fezes/microbiologia , Técnicas de Diagnóstico Molecular/métodos , Adulto , Humanos , Limite de Detecção
11.
Res Sq ; 2023 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-37609264

RESUMO

Background: It is estimated that more than half of the world population has been infected with Helicobacter pylori. Most newly acquired H. pylori infections occur in children before 10 years of age. We hypothesized that early life H. pylori infection could influence the composition of the microbiome at mucosal sites distant to the stomach. To test this hypothesis, we utilized the infant rhesus macaque monkey as an animal model of natural H. pylori colonization to determine the impact of infection on the lung and oral microbiome during a window of postnatal development. Results: From a cohort of 4-7-month-old monkeys, gastric biopsy cultures identified 44% of animals infected by H. pylori. 16S ribosomal RNA gene sequencing of lung washes and buccal swabs from animals showed distinct profiles for the lung and oral microbiome, independent of H. pylori infection. In relative order of abundance, the lung microbiome was dominated by the phyla Proteobacteria, Firmicutes, Bacteroidota, Fusobacteriota, Campilobacterota and Actinobacteriota while the oral microbiome was dominated by Proteobacteria, Firmicutes, Bacteroidota, and Fusobacteriota. Relative to the oral cavity, the lung was composed of more genera and species that significantly differed by H. pylori status, with a total of 6 genera and species that were increased in H. pylori negative infant monkey lungs. Lung, but not plasma IL-8 concentration was also associated with gastric H. pylori load and lung microbial composition. Conclusions: We found the infant rhesus macaque monkey lung harbors a microbiome signature that is distinct from that of the oral cavity during postnatal development. Gastric H. pylori colonization and IL-8 protein were linked to the composition of microbial communities in the lung and oral cavity. Collectively, these findings provide insight into how H. pylori infection might contribute to the gut-lung axis during early childhood and modulate future respiratory health.

12.
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.

13.
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
14.
Clin Infect Dis ; 55(7): 982-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22700831

RESUMO

Diarrhea is common among hospitalized patients but the causes are distinct from those of diarrhea in the community. We review existing data about the epidemiology of nosocomial diarrhea and summarize recent progress in understanding the mechanisms of diarrhea. Clinicians should recognize that most cases of nosocomial diarrhea have a noninfectious etiology, including medications, underlying illness, and enteral feeding. Apart from Clostridium difficile, the frequency of infectious causes such as norovirus and toxigenic strains of Clostridium perfringens, Klebsiella oxytoca, Staphylococcus aureus, and Bacteroides fragilis remains largely undefined and test availability is limited. Here we provide a practical approach to the evaluation and management of nosocomial diarrhea when tests for C. difficile are negative.


Assuntos
Infecções Bacterianas/epidemiologia , Infecção Hospitalar/epidemiologia , Infecção Hospitalar/etiologia , Diarreia/epidemiologia , Diarreia/etiologia , Viroses/epidemiologia , Infecções Bacterianas/microbiologia , Infecção Hospitalar/diagnóstico , Infecção Hospitalar/terapia , Diarreia/diagnóstico , Diarreia/terapia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/terapia , Humanos , Viroses/virologia
15.
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
16.
Helicobacter ; 16 Suppl 1: 26-32, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21896082

RESUMO

The immune response to Helicobacter pylori entails both innate effectors and a complex mix of Th1, Th17, and Treg adaptive immune responses. The clinical outcome of infection may well depend to a large degree on the relative balance of these responses. Vaccination with a wide range of antigens, adjuvants, and delivery routes can produce statistically significant reductions in H. pylori colonization levels in mice, though rarely sterilizing immunity. Whether similar reductions in bacterial load can be achieved in humans, and whether they would be clinically significant, is still unclear. However, progress in understanding the role of Th1, Th17, and most recently Treg cells in protection against H. pylori infection provides reason for optimism.


Assuntos
Vacinas Bacterianas/imunologia , Desenho de Fármacos , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Imunidade , Animais , Vacinas Bacterianas/genética , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/genética , Humanos , Camundongos
17.
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
19.
Gastroenterology ; 137(3): 1061-71, 1071.e1-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19375420

RESUMO

BACKGROUND & AIMS: Helicobacter pylori attaches to mucin oligosaccharides that are expressed on host gastric epithelium. We used the rhesus macaque model to characterize the effect of H. pylori infection on gastric mucin oligosaccharides during acute and chronic infection. METHODS: Specific pathogen (H. pylori)-free rhesus macaques were inoculated with H. pylori J166. Biopsy specimens of the gastric antrum were obtained 2 and 4 weeks before and 2, 8, and 24 weeks after infection with H. pylori. O-linked mucin oligosaccharides were released from gastric biopsy samples by beta-elimination and profiled by matrix-assisted laser desorption/ionization mass spectrometry. Similar studies were performed on gastric biopsy samples from H. pylori-infected and uninfected humans. Formalin-fixed, paraffin-embedded sections of rhesus antrum biopsy samples were stained with H&E, periodic acid-Schiff stain, and antibody to MUC5AC, the predominant mucin expressed in the stomach. RESULTS: H. pylori-induced gastritis was accompanied by an acute and dramatic decrease in diversity and relative abundance of O-linked mucin oligosaccharides in the rhesus stomach, which largely recovered during the 24-week observation period. These variations in oligosaccharide abundance detected by mass spectrometry were reflected by changes in periodic acid-Schiff-positive material and expression of MUC5AC over time. Relatively few differences were seen in gastric mucin oligosaccharide composition between H. pylori-infected and uninfected patients, which is consistent with the results in rhesus macaques because infection occurs in childhood. CONCLUSIONS: Acute H. pylori infection is accompanied by a dramatic but transient loss in mucin oligosaccharides that may promote colonization and persistence.


Assuntos
Mucinas Gástricas/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori , Oligossacarídeos/metabolismo , Animais , Gastrite/metabolismo , Gastrite/microbiologia , Gastrite/patologia , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/isolamento & purificação , Macaca mulatta , Masculino , Mucina-5AC/metabolismo , Antro Pilórico/microbiologia , Antro Pilórico/patologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA