Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Metab ; 34(11): 1779-1791.e9, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36240759

RESUMO

Microbiome dysbiosis is a feature of diabetes, but how microbial products influence insulin production is poorly understood. We report the mechanism of BefA, a microbiome-derived protein that increases proliferation of insulin-producing ß cells during development in gnotobiotic zebrafish and mice. BefA disseminates systemically by multiple anatomic routes to act directly on pancreatic islets. We detail BefA's atomic structure, containing a lipid-binding SYLF domain, and demonstrate that it permeabilizes synthetic liposomes and bacterial membranes. A BefA mutant impaired in membrane disruption fails to expand ß cells, whereas the pore-forming host defense protein, Reg3, stimulates ß cell proliferation. Our work demonstrates that membrane permeabilization by microbiome-derived and host defense proteins is necessary and sufficient for ß cell expansion during pancreas development, potentially connecting microbiome composition with diabetes risk.


Assuntos
Diabetes Mellitus , Microbiota , Camundongos , Animais , Peixe-Zebra , Pâncreas/metabolismo , Insulina/metabolismo , Diabetes Mellitus/metabolismo , Proteínas/metabolismo
2.
mBio ; 12(4): e0181921, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34340539

RESUMO

The Helicobacter pylori chemoreceptor TlpA plays a role in dampening host inflammation during chronic stomach colonization. TlpA has a periplasmic dCache_1 domain, a structure that is capable of sensing many ligands; however, the only characterized TlpA signals are arginine, bicarbonate, and acid. To increase our understanding of TlpA's sensing profile, we screened for diverse TlpA ligands using ligand binding arrays. TlpA bound seven ligands with affinities in the low- to middle-micromolar ranges. Three of these ligands, arginine, fumarate, and cysteine, were TlpA-dependent chemoattractants, while the others elicited no response. Molecular docking experiments, site-directed point mutants, and competition surface plasmon resonance binding assays suggested that TlpA binds ligands via both the membrane-distal and -proximal dCache_1 binding pockets. Surprisingly, one of the nonactive ligands, glucosamine, acted as a chemotaxis antagonist, preventing the chemotaxis response to chemoattractant ligands, and acted to block the binding of ligands irrespective of whether they bound the membrane-distal or -proximal dCache_1 subdomains. In total, these results suggest that TlpA senses multiple attractant ligands as well as antagonist ones, an emerging theme in chemotaxis systems. IMPORTANCE Numerous chemotactic bacterial pathogens depend on the ability to sense a diverse array of signals through chemoreceptors to achieve successful colonization and virulence within their host. The signals sensed by chemoreceptors, however, are not always fully understood. This is the case for TlpA, a dCache_1 chemoreceptor of H. pylori that enables the bacterium to induce less inflammation during chronic infections. H. pylori causes a significant global disease burden, which is driven by the development of gastric inflammation. Accordingly, it is essential to understand the processes by which H. pylori modulates host inflammation. This work uncovers the signals that TlpA can sense and highlights the underappreciated ability to regulate chemotactic responses by antagonistic chemoreceptor ligands, which is an emerging theme among other chemotactic systems.


Assuntos
Proteínas de Bactérias/metabolismo , Células Quimiorreceptoras/metabolismo , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Proteínas de Bactérias/genética , Quimiotaxia , Glucosamina/metabolismo , Ligantes , Simulação de Acoplamento Molecular , Mutação Puntual
3.
Elife ; 72018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30398151

RESUMO

Stable mutualism between a host and its resident bacteria requires a moderated immune response to control bacterial population size without eliciting excessive inflammation that could harm both partners. Little is known about the specific molecular mechanisms utilized by bacterial mutualists to temper their hosts' responses and protect themselves from aggressive immune attack. Using a gnotobiotic larval zebrafish model, we identified an Aeromonas secreted immunomodulatory protein, AimA. AimA is required during colonization to prevent intestinal inflammation that simultaneously compromises both bacterial and host survival. Administration of exogenous AimA prevents excessive intestinal neutrophil accumulation and protects against septic shock in models of both bacterially and chemically induced intestinal inflammation. We determined the molecular structure of AimA, which revealed two related calycin-like domains with structural similarity to the mammalian immune modulatory protein, lipocalin-2. As a secreted bacterial protein required by both partners for optimal fitness, AimA is an exemplar bacterial mutualism factor.


Assuntos
Aeromonas/genética , Interações Hospedeiro-Patógeno/genética , Imunidade Inata/genética , Simbiose/genética , Animais , Interações Hospedeiro-Patógeno/imunologia , Intestinos/imunologia , Intestinos/microbiologia , Larva/imunologia , Larva/microbiologia , Lipocalinas/genética , Lipocalinas/imunologia , Domínios Proteicos/genética , Simbiose/imunologia , Peixe-Zebra/imunologia , Peixe-Zebra/microbiologia
4.
Cell Host Microbe ; 19(1): 5-7, 2016 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-26764590

RESUMO

Long before the cause was discovered, peptic ulcers were known to occur preferentially in individuals of certain blood groups. In this issue of Cell Host & Microbe, Moonens et al. (2016) perform structural analyses of the Helicobacter pylori adhesin BabA to determine how the bacteria discriminately bind different blood group glycans.


Assuntos
Infecções por Helicobacter/microbiologia , Helicobacter pylori/metabolismo , Adesinas Bacterianas/metabolismo , Humanos
5.
Cell Host Microbe ; 18(2): 147-56, 2015 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-26269952

RESUMO

The gastric pathogen Helicobacter pylori interacts intimately with the gastric mucosa to avoid the microbicidal acid in the stomach lumen. The cues H. pylori senses to locate and colonize the gastric epithelium have not been well defined. We show that metabolites emanating from human gastric organoids rapidly attract H. pylori. This response is largely controlled by the bacterial chemoreceptor TlpB, and the main attractant emanating from epithelia is urea. Our previous structural analyses show that TlpB binds urea with high affinity. Here we demonstrate that this tight binding controls highly sensitive responses, allowing detection of urea concentrations as low as 50 nM. Attraction to urea requires that H. pylori urease simultaneously destroys the signal. We propose that H. pylori has evolved a sensitive urea chemodetection and destruction system that allows the bacterium to dynamically and locally modify the host environment to locate the epithelium.


Assuntos
Proteínas de Bactérias/metabolismo , Quimiotaxia , Epitélio/microbiologia , Mucosa Gástrica/microbiologia , Helicobacter pylori/fisiologia , Ureia/metabolismo , Urease/metabolismo , Animais , Modelos Animais de Doenças , Epitélio/metabolismo , Mucosa Gástrica/metabolismo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos
6.
mBio ; 6(4): e00379, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26152582

RESUMO

UNLABELLED: The gastric pathogen Helicobacter pylori forms biofilms on abiotic and biotic surfaces. We have shown previously that H. pylori perceives the quorum signal autoinducer-2 (AI-2) as a chemorepellent. We report here that H. pylori chemorepulsion from endogenous AI-2 influences the proportions and spatial organization of cells within biofilms. Strains that fail to produce AI-2 (∆luxS strains) or are defective for chemotaxis (∆cheA strains) formed more spatially homogenous biofilms with a greater proportion of adherent versus planktonic cells than wild-type biofilms. Reciprocally, a strain that overproduced AI-2 (luxS(OP)) formed biofilms with proportionally fewer adherent cells. Along with the known AI-2 chemoreceptor, TlpB, we identified AibA and AibB, two novel periplasmic binding proteins that are required for the AI-2 chemorepulsion response. Disruptions in any of the proteins required for AI-2 chemotaxis recapitulated the biofilm adherence and spatial organization phenotype of the ∆luxS mutant. Furthermore, exogenous administration of AI-2 was sufficient to decrease the proportion of adherent cells in biofilms and promote dispersal of cells from biofilms in a chemotaxis-dependent manner. Finally, we found that disruption of AI-2 production or AI-2 chemotaxis resulted in increased clustering of cells in microcolonies on cultured epithelial cells. We conclude that chemotaxis from AI-2 is a determinant of H. pylori biofilm spatial organization and dispersal. IMPORTANCE: Bacterial biofilms are ubiquitous in nature, but the mechanisms governing their assembly and spatial organization are not fully understood. Bacterial communication through quorum sensing has been shown to influence biofilm growth through the regulation of biofilm genes. Our study revealed a new role for quorum sensing in biofilms through rapid chemotactic responses to quorum signals. Specifically, we studied how chemorepulsion of Helicobacter pylori from the universal quorum signal autoinducer-2 (AI-2) shapes the spatial organization of its biofilms. We demonstrate that the chemorepulsive response of H. pylori to AI-2 is necessary to promote its dispersal from biofilms grown on both abiotic and biotic surfaces and is sufficient to promote dispersal in a chemotaxis-dependent manner. This work has broad implications for understanding the mechanisms by which endogenously produced microbial compounds shape the assembly and spatial organization of microbial communities in their environments.


Assuntos
Biofilmes/crescimento & desenvolvimento , Quimiotaxia , Helicobacter pylori/fisiologia , Homosserina/análogos & derivados , Lactonas/metabolismo , Proteínas de Bactérias/metabolismo , Liases de Carbono-Enxofre/deficiência , Liases de Carbono-Enxofre/metabolismo , Deleção de Genes , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Homosserina/metabolismo , Proteínas de Membrana/deficiência , Proteínas de Membrana/metabolismo , Proteínas Quimiotáticas Aceptoras de Metil , Proteínas Periplásmicas/genética , Proteínas Periplásmicas/metabolismo
7.
mBio ; 2(5)2011.
Artigo em Inglês | MEDLINE | ID: mdl-21933915

RESUMO

For almost 50 years, Escherichia coli has been the model for understanding how bacteria orient their movement in response to chemical cues, but recent studies of chemotaxis in other bacteria have revealed interesting variations from prevailing paradigms. Investigating the human pathogen Helicobacter pylori, Amieva and colleagues [mBio 2(4):e00098-11, 2011] discovered a new chemotaxis regulator, ChePep, which modulates swimming behavior through the canonical histidine-aspartate phosphorelay system. Functionally conserved among the epsilonproteobacteria, ChePep is essential for H. pylori to navigate deep into the stomach's gastric glands and may be an attractive target for novel antibiotics.


Assuntos
Proteínas de Bactérias/metabolismo , Quimiotaxia , Epsilonproteobacteria/fisiologia , Epsilonproteobacteria/patogenicidade , Mucosa Gástrica/microbiologia , Fatores de Virulência/metabolismo , Animais , Feminino
8.
Microbiology (Reading) ; 157(Pt 9): 2445-2455, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21602215

RESUMO

Helicobacter pylori moves in response to environmental chemical cues using a chemotaxis two-component signal-transduction system. Autoinducer-2 (AI-2) is a quorum-sensing signal produced by the LuxS protein that accumulates in the bacterial environment in a density-dependent manner. We showed previously that a H. pylori luxS mutant was defective in motility on soft agar plates. Here we report that deletion of the luxS gene resulted in swimming behaviour with a reduced frequency of stops as compared to the wild-type strain. Stopping frequency was restored to wild-type levels by genetic complementation of the luxS mutation or by addition of synthetic 4,5-dihydroxy-2,3-pentanedione (DPD), which cyclizes to form AI-2. Synthetic DPD also increased the frequency of stops in wild-type H. pylori, similar to the behaviour induced by the known chemorepellent HCl. We found that whereas mutants lacking the chemoreceptor genes tlpA, tlpC or tlpD responded to an exogenous source of synthetic DPD, the chemoreceptor mutant tlpB was non-responsive to a gradient or uniform distribution of the chemical. Furthermore, a double mutant lacking both tlpB and luxS exhibited chemotactic behaviour similar to the tlpB single mutant, whereas a double mutant lacking both tlpB and the chemotransduction gene cheA behaved like a nonchemotactic cheA single mutant, supporting the model that tlpB functions in a signalling pathway downstream of luxS and upstream of cheA. We conclude that H. pylori perceives LuxS-produced AI-2 as a chemorepellent via the chemoreceptor TlpB.


Assuntos
Proteínas de Bactérias/metabolismo , Helicobacter pylori/metabolismo , Homosserina/análogos & derivados , Lactonas/metabolismo , Percepção de Quorum , Proteínas de Bactérias/genética , Liases de Carbono-Enxofre/genética , Liases de Carbono-Enxofre/metabolismo , Quimiotaxia/efeitos dos fármacos , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/genética , Homosserina/metabolismo , Pentanos/metabolismo , Pentanos/farmacologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...