Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
PLoS Pathog ; 15(9): e1007972, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31487328

RESUMEN

The biogenesis of bacterial cell-envelope polysaccharides requires the translocation, across the plasma membrane, of sugar sub-units that are produced inside the cytoplasm. To this end, the hydrophilic sugars are anchored to a lipid phosphate carrier (undecaprenyl phosphate (C55-P)), yielding membrane intermediates which are translocated to the outer face of the membrane. Finally, the glycan moiety is transferred to a nascent acceptor polymer, releasing the carrier in the "inactive" undecaprenyl pyrophosphate (C55-PP) form. Thus, C55-P is generated through the dephosphorylation of C55-PP, itself arising from either de novo synthesis or recycling. Two types of integral membrane C55-PP phosphatases were described: BacA enzymes and a sub-group of PAP2 enzymes (type 2 phosphatidic acid phosphatases). The human pathogen Helicobacter pylori does not contain BacA homologue but has four membrane PAP2 proteins: LpxE, LpxF, HP0350 and HP0851. Here, we report the physiological role of HP0851, renamed HupA, via multiple and complementary approaches ranging from a detailed biochemical characterization to the assessment of its effect on cell envelope metabolism and microbe-host interactions. HupA displays a dual function as being the main C55-PP pyrophosphatase (UppP) and phosphatidylglycerol phosphate phosphatase (PGPase). Although not essential in vitro, HupA was essential in vivo for stomach colonization. In vitro, the remaining UppP activity was carried out by LpxE in addition to its lipid A 1-phosphate phosphatase activity. Both HupA and LpxE have crucial roles in the biosynthesis of several cell wall polysaccharides and thus constitute potential targets for new therapeutic strategies.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Helicobacter pylori/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de la Membrana Bacteriana Externa/fisiología , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Pared Celular/metabolismo , Proteínas de Unión al ADN , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Femenino , Helicobacter pylori/patogenicidad , Ratones , Ratones Endogámicos , Pruebas de Sensibilidad Microbiana , Fosfatidato Fosfatasa , Monoéster Fosfórico Hidrolasas/metabolismo , Fosfatos de Poliisoprenilo/metabolismo , Polimixina B/farmacología , Pirofosfatasas/metabolismo , Estómago
2.
J Infect Dis ; 210(8): 1286-95, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24755437

RESUMEN

Adhesion of Helicobacter pylori to the gastric mucosa is a necessary prerequisite for the pathogenesis of H. pylori-related diseases. In this study, we investigated the GalNAcß1-4GlcNAc motif (also known as N,N'-diacetyllactosediamine [lacdiNAc]) carried by MUC5AC gastric mucins as the target for bacterial binding to the human gastric mucosa. The expression of LacdiNAc carried by gastric mucins was correlated with H. pylori localization, and all strains tested adhered significantly to this motif. Proteomic analysis and mutant construction allowed the identification of a yet uncharacterized bacterial adhesin, LabA, which specifically recognizes lacdiNAc. These findings unravel a target of adhesion for H. pylori in addition to moieties recognized by the well-characterized adhesins BabA and SabA. Localization of the LabA target, restricted to the gastric mucosa, suggests a plausible explanation for the tissue tropism of these bacteria. These results pave the way for the development of alternative strategies against H. pylori infection, using adherence inhibitors.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana/fisiología , Mucosa Gástrica/microbiología , Regulación Bacteriana de la Expresión Génica/fisiología , Helicobacter pylori/fisiología , Adhesinas Bacterianas/genética , Secuencia de Aminoácidos , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Unión Proteica , Ratas , Ratas Sprague-Dawley
3.
Mol Microbiol ; 86(4): 845-56, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22994973

RESUMEN

The flagellar machinery is a highly complex organelle composed of a free rotating flagellum and a fixed stator that converts energy into movement. The assembly of the flagella and the stator requires interactions with the peptidoglycan layer through which the organelle has to pass for externalization. Lytic transglycosylases are peptidoglycan degrading enzymes that cleave the sugar backbone of peptidoglycan layer. We show that an endogenous lytic transglycosylase is required for full motility of Helicobacter pylori and colonization of the gastric mucosa. Deficiency of motility resulted from a paralysed phenotype implying an altered ability to generate flagellar rotation. Similarly, another Gram-negative pathogen Salmonella typhimurium and the Gram-positive pathogen Listeria monocytogenes required the activity of lytic transglycosylases, Slt or MltC, and a glucosaminidase (Auto), respectively, for full motility. Furthermore, we show that in absence of the appropriate lytic transglycosylase, the flagellar motor protein MotB from H. pylori does not localize properly to the bacterial pole. We present a new model involving the maturation of the surrounding peptidoglycan for the proper anchoring and functionality of the flagellar motor.


Asunto(s)
Flagelos/fisiología , Glicosiltransferasas/metabolismo , Helicobacter pylori/enzimología , Hexosaminidasas/metabolismo , Listeria monocytogenes/enzimología , Peptidoglicano/metabolismo , Salmonella typhimurium/enzimología , Helicobacter pylori/fisiología , Listeria monocytogenes/fisiología , Sustancias Macromoleculares/metabolismo , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Proteínas Motoras Moleculares/metabolismo , Transporte de Proteínas , Salmonella typhimurium/fisiología
4.
PLoS Pathog ; 7(12): e1002454, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22216004

RESUMEN

Modification of bacterial surface structures, such as the lipid A portion of lipopolysaccharide (LPS), is used by many pathogenic bacteria to help evade the host innate immune response. Helicobacter pylori, a gram-negative bacterium capable of chronic colonization of the human stomach, modifies its lipid A by removal of phosphate groups from the 1- and 4'-positions of the lipid A backbone. In this study, we identify the enzyme responsible for dephosphorylation of the lipid A 4'-phosphate group in H. pylori, Jhp1487 (LpxF). To ascertain the role these modifications play in the pathogenesis of H. pylori, we created mutants in lpxE (1-phosphatase), lpxF (4'-phosphatase) and a double lpxE/F mutant. Analysis of lipid A isolated from lpxE and lpxF mutants revealed lipid A species with a 1 or 4'-phosphate group, respectively while the double lpxE/F mutant revealed a bis-phosphorylated lipid A. Mutants lacking lpxE, lpxF, or lpxE/F show a 16, 360 and 1020 fold increase in sensitivity to the cationic antimicrobial peptide polymyxin B, respectively. Moreover, a similar loss of resistance is seen against a variety of CAMPs found in the human body including LL37, ß-defensin 2, and P-113. Using a fluorescent derivative of polymyxin we demonstrate that, unlike wild type bacteria, polymyxin readily associates with the lpxE/F mutant. Presumably, the increase in the negative charge of H. pylori LPS allows for binding of the peptide to the bacterial surface. Interestingly, the action of LpxE and LpxF was shown to decrease recognition of Helicobacter LPS by the innate immune receptor, Toll-like Receptor 4. Furthermore, lpxE/F mutants were unable to colonize the gastric mucosa of C57BL/6J and C57BL/6J tlr4 -/- mice when compared to wild type H. pylori. Our results demonstrate that dephosphorylation of the lipid A domain of H. pylori LPS by LpxE and LpxF is key to its ability to colonize a mammalian host.


Asunto(s)
Membrana Celular/metabolismo , Mucosa Gástrica/microbiología , Helicobacter pylori/fisiología , Interacciones Huésped-Patógeno/fisiología , Inmunidad Innata/fisiología , Lípido A/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/inmunología , Péptidos Catiónicos Antimicrobianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Membrana Celular/genética , Membrana Celular/inmunología , Mucosa Gástrica/inmunología , Células HEK293 , Humanos , Lípido A/genética , Lípido A/inmunología , Ratones , Ratones Noqueados , Mutación , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/inmunología , Monoéster Fosfórico Hidrolasas/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo
5.
Glycobiology ; 22(9): 1193-206, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22522599

RESUMEN

Helicobacter pylori infects more than half of the world's population. Although most patients are asymptomatic, persistent infection may cause chronic gastritis and gastric cancer. Adhesion of the bacteria to the gastric mucosa is a necessary prerequisite for the pathogenesis of H. pylori-related diseases and is mediated by mucin O-glycans. In order to define which glycans may be implicated in the binding of the bacteria to the gastric mucosa in humans, we have characterized the exact pattern of glycosylation of gastric mucins. We have identified that the major component was always a core 2-based glycan carrying two blood group H antigens, whatever was the blood group of individuals. We have also demonstrated that around 80% of O-glycans carried blood group A, B or H antigens, suggesting that the variation of gastric mucin glycosylation between individuals is partly due to the blood group status. This study will help better understanding the role of O-glycans in the physiology and homeostasis of gastric mucosa. Overall, the results reported here give us the necessary background information to begin studies to determine whether individuals who express certain carbohydrate epitopes on specific mucins are predisposed to certain gastric diseases.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/química , Mucinas Gástricas/química , Mucosa Gástrica/química , Helicobacter pylori/química , Antígenos del Grupo Sanguíneo de Lewis/química , Polisacáridos/química , Sistema del Grupo Sanguíneo ABO/metabolismo , Adolescente , Adulto , Sitios de Unión , Secuencia de Carbohidratos , Cromatografía Líquida de Alta Presión , Susceptibilidad a Enfermedades , Femenino , Mucinas Gástricas/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Glicosilación , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/microbiología , Helicobacter pylori/metabolismo , Humanos , Antígenos del Grupo Sanguíneo de Lewis/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Datos de Secuencia Molecular , Polisacáridos/metabolismo , Unión Proteica
6.
Mol Microbiol ; 82(1): 68-86, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21801243

RESUMEN

The definition of bacterial cell shape is a complex process requiring the participation of multiple components of an intricate macromolecular machinery. We aimed at characterizing the determinants involved in cell shape of the helical bacterium Helicobacter pylori. Using a yeast two-hybrid screen with the key cell elongation protein PBP2 as bait, we identified an interaction between PBP2 and MreC. The minimal region of MreC required for this interaction ranges from amino acids 116 to 226. Using recombinant proteins, we showed by affinity and size exclusion chromatographies and surface plasmon resonance that PBP2 and MreC form a stable complex. In vivo, the two proteins display a similar spatial localization and their complex has an apparent 1:1 stoichiometry; these results were confirmed in vitro by analytical ultracentrifugation and chemical cross-linking. Small angle X-ray scattering analyses of the PBP2 : MreC complex suggest that MreC interacts directly with the C-terminal region of PBP2. Depletion of either PBP2 or MreC leads to transition into spherical cells that lose viability. Finally, the specific expression in trans of the minimal interacting domain of MreC with PBP2 in the periplasmic space leads to cell rounding, suggesting that the PBP2/MreC complex formation in vivo is essential for cell morphology.


Asunto(s)
Proteínas Bacterianas/metabolismo , Helicobacter pylori/citología , Helicobacter pylori/metabolismo , Proteínas de Unión a las Penicilinas/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Helicobacter pylori/química , Helicobacter pylori/genética , Viabilidad Microbiana , Datos de Secuencia Molecular , Proteínas de Unión a las Penicilinas/química , Proteínas de Unión a las Penicilinas/genética , Unión Proteica , Estructura Terciaria de Proteína , Alineación de Secuencia , Técnicas del Sistema de Dos Híbridos
7.
Mol Microbiol ; 78(4): 809-19, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20815828

RESUMEN

The molecular basis of the regulation of specific shapes and their role for the bacterial fitness remain largely unknown. We focused in this study on the Gram-negative and spiral-shaped Helicobacter pylori. To colonize its unique niche, H. pylori needs to reach quickly the human gastric mucosa, by swimming to and through the mucus layer. For that reason, the specific shape of H. pylori is predicted to be necessary for optimal motility in vivo, and consequently for its colonization ability. Here, we describe the involvement of a PG-modifying enzyme, HdpA (HP0506), in the mouse colonization ability of this bacterium, by regulating its shape. Indeed, the inactivation of the hp0506 gene led to a stocky and branched phenotype, affecting H. pylori colonization capacity despite a normal motility phenotype in vitro. In contrast, the overexpression of the hp0506 gene induced the transformation of H. pylori from rod to dividing cocci shaped bacteria. Furthermore, we demonstrated by PG analysis and enzymology, that HdpA carried both d,d-carboxypeptidase and d,d-endopeptidase activities. Thus, HdpA is the first enzyme belonging to the M23-peptidase family able to perform the d,d-carboxypeptidation and regulate cell shape.


Asunto(s)
Proteínas Bacterianas/metabolismo , Helicobacter pylori/citología , Helicobacter pylori/patogenicidad , Metaloproteasas/metabolismo , Peptidoglicano/metabolismo , Factores de Virulencia/metabolismo , Animales , Carboxipeptidasas/genética , Carboxipeptidasas/metabolismo , Pared Celular/metabolismo , Endopeptidasas/genética , Endopeptidasas/metabolismo , Técnicas de Inactivación de Genes , Helicobacter pylori/enzimología , Metaloproteasas/genética , Ratones
8.
BMC Microbiol ; 10: 91, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20346161

RESUMEN

BACKGROUND: Trans-translation is a ubiquitous bacterial quality control-mechanism for both transcription and translation. With its two major partners, SsrA a small stable RNA and the SmpB protein, it promotes the release of ribosomes stalled on defective mRNAs and directs the corresponding truncated proteins to degradation pathways. We have recently shown that trans-translation is an essential function in the gastric pathogen Helicobacter pylori. Our results suggested that some properties of the H. pylori trans-translation machinery distinguishes it from the well known system in E. coli. Therefore, we decided to test the functionality of the SmpB and SsrA molecules of H. pylori in the E. coli heterologous system using two established phenotypic tests. RESULTS: H. pylori SmpB protein was found to successfully restore the E. coli DeltasmpB mutant growth defect and its capacity to propagate lambdaimmP22 phage. We showed that in E. coli, H. pylori SsrA (Hp-SsrA) was stably expressed and maturated and that this molecule could restore wild type growth to the E. coli DeltassrA mutant. Hp-SsrA mutants affected in the ribosome rescue function were not able to restore normal growth to E. coli DeltassrA supporting a major role of ribosome rescue in this phenotype. Surprisingly, Hp-SsrA did not restore the phage lambdaimmP22 propagation capacity to the E. coli DeltassrA mutant. CONCLUSIONS: These data suggest an additional role of the tag sequence that presents specific features in Hp-SsrA. Our interpretation is that a secondary role of protein tagging in phage propagation is revealed by heterologous complementation because ribosome rescue is less efficient. In conclusion, tmRNAs present in all eubacteria, have coevolved with the translational machinery of their host and possess specific determinants that can be revealed by heterologous complementation studies.


Asunto(s)
Prueba de Complementación Genética , Helicobacter pylori/fisiología , Biosíntesis de Proteínas , ARN Bacteriano/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcripción Genética , Bacteriófago lambda/crecimiento & desarrollo , Escherichia coli/genética , Eliminación de Gen , Helicobacter pylori/genética , Viabilidad Microbiana , ARN Bacteriano/genética , Proteínas de Unión al ARN/genética
9.
Mol Cell Proteomics ; 7(12): 2429-41, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18682379

RESUMEN

In the human gastric bacterium Helicobacter pylori, two metalloenzymes, hydrogenase and urease, are essential for in vivo colonization, the latter being a major virulence factor. The UreA and UreB structural subunits of urease and UreG, one of the accessory proteins for Ni(2+) incorporation into apourease, were taken as baits for tandem affinity purification. The method allows the purification of protein complexes under native conditions and physiological expression levels of the bait protein. Furthermore the tandem affinity purification technology was combined with in vivo cross-link to capture transient interactions. The results revealed different populations of urease complexes: (i) urease captured during activation by Ni(2+) ions comprising all the accessory proteins and (ii) urease in association with metabolic proteins involved e.g. in ammonium incorporation and the cytoskeleton. Using UreG as a bait protein, we copurified HypB, the accessory protein for Ni(2+) incorporation into hydrogenase, that is reported to play a role in urease activation. The interactome of HypB partially overlapped with that of urease and revealed interactions with SlyD, which is known to be involved in hydrogenase maturation as well as with proteins implicated in the formation of [Fe-S] clusters present in the small subunit of hydrogenase. In conclusion, this study provides new insight into coupling of ammonium production and assimilation in the gastric pathogen and the intimate link between urease and hydrogenase maturation.


Asunto(s)
Cromatografía de Afinidad/métodos , Helicobacter pylori/enzimología , Ureasa/metabolismo , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Cromosomas Bacterianos , Mezclas Complejas/metabolismo , Reactivos de Enlaces Cruzados/farmacología , Estudios de Factibilidad , Glutamato-Amoníaco Ligasa/metabolismo , Helicobacter pylori/efectos de los fármacos , Hidrogenasas/metabolismo , Immunoblotting , Inmunoglobulina G/metabolismo , Proteínas Hierro-Azufre/metabolismo , Metales/metabolismo , Modelos Biológicos , Nitrógeno/metabolismo , Unión Proteica/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Recombinación Genética/genética , Urea/metabolismo
10.
Infect Immun ; 76(2): 497-509, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18025096

RESUMEN

Carbon dioxide occupies a central position in the physiology of Helicobacter pylori owing to its capnophilic nature, the large amounts of carbon dioxide produced by urease-mediated urea hydrolysis, and the constant bicarbonate supply in the stomach. Carbonic anhydrases (CA) catalyze the interconversion of carbon dioxide and bicarbonate and are involved in functions such as CO(2) transport or trapping and pH homeostasis. H. pylori encodes a periplasmic alpha-CA (alpha-CA-HP) and a cytoplasmic beta-CA (beta-CA-HP). Single CA inactivation and double CA inactivation were obtained for five genetic backgrounds, indicating that H. pylori CA are not essential for growth in vitro. Bicarbonate-carbon dioxide exchange rates were measured by nuclear magnetic resonance spectroscopy using lysates of parental strains and CA mutants. Only the mutants defective in the alpha-CA-HP enzyme showed strongly reduced exchange rates. In H. pylori, urease activity is essential for acid resistance in the gastric environment. Urease activity measured using crude cell extracts was not modified by the absence of CA. With intact CA mutant cells incubated in acidic conditions (pH 2.2) in the presence of urea there was a delay in the increase in the pH of the incubation medium, a phenotype most pronounced in the absence of H. pylori alpha-CA. This correlated with a delay in acid activation of the urease as measured by slower ammonia production in whole cells. The role of CA in vivo was examined using the mouse model of infection with two mouse-adapted H. pylori strains, SS1 and X47-2AL. Compared to colonization by the wild-type strain, colonization by X47-2AL single and double CA mutants was strongly reduced. Colonization by SS1 CA mutants was not significantly different from colonization by wild-type strain SS1. However, when mice were infected by SS1 Delta(beta-CA-HP) or by a SS1 double CA mutant, the inflammation scores of the mouse gastric mucosa were strongly reduced. In conclusion, CA contribute to the urease-dependent response to acidity of H. pylori and are required for high-grade inflammation and efficient colonization by some strains.


Asunto(s)
Ácidos/metabolismo , Proteínas Bacterianas/metabolismo , Anhidrasas Carbónicas/metabolismo , Mucosa Gástrica/microbiología , Helicobacter pylori/patogenicidad , Ureasa/metabolismo , Amoníaco/metabolismo , Animales , Proteínas Bacterianas/genética , Bicarbonatos/metabolismo , Dióxido de Carbono/metabolismo , Anhidrasas Carbónicas/genética , Recuento de Colonia Microbiana , Femenino , Eliminación de Gen , Helicobacter pylori/enzimología , Espectroscopía de Resonancia Magnética , Ratones , Mutagénesis Insercional , Ureasa/genética
11.
PLoS Pathog ; 2(9): e97, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17002496

RESUMEN

The human gastric pathogen Helicobacter pylori is responsible for peptic ulcers and neoplasia. Both in vitro and in the human stomach it can be found in two forms, the bacillary and coccoid forms. The molecular mechanisms of the morphological transition between these two forms and the role of coccoids remain largely unknown. The peptidoglycan (PG) layer is a major determinant of bacterial cell shape, and therefore we studied H. pylori PG structure during the morphological transition. The transition correlated with an accumulation of the N-acetyl-D-glucosaminyl-beta(1,4)-N-acetylmuramyl-L-Ala-D-Glu (GM-dipeptide) motif. We investigated the molecular mechanisms responsible for the GM-dipeptide motif accumulation, and studied the role of various putative PG hydrolases in this process. Interestingly, a mutant strain with a mutation in the amiA gene, encoding a putative PG hydrolase, was impaired in accumulating the GM-dipeptide motif and transforming into coccoids. We investigated the role of the morphological transition and the PG modification in the biology of H. pylori. PG modification and transformation of H. pylori was accompanied by an escape from detection by human Nod1 and the absence of NF-kappaB activation in epithelial cells. Accordingly, coccoids were unable to induce IL-8 secretion by AGS gastric epithelial cells. amiA is, to our knowledge, the first genetic determinant discovered to be required for this morphological transition into the coccoid forms, and therefore contributes to modulation of the host response and participates in the chronicity of H. pylori infection.


Asunto(s)
Proteínas Bacterianas/fisiología , Proteínas Portadoras/fisiología , Helicobacter pylori/fisiología , Lipoproteínas/fisiología , Secuencias de Aminoácidos , Amoxicilina/farmacología , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Proteínas Portadoras/genética , Línea Celular , Forma de la Célula/fisiología , Pared Celular/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/fisiología , Prueba de Complementación Genética , Helicobacter pylori/citología , Helicobacter pylori/genética , Helicobacter pylori/inmunología , Humanos , Lipoproteínas/genética , Mutación , Peptidoglicano/genética , Peptidoglicano/metabolismo
12.
Appl Environ Microbiol ; 74(7): 2095-102, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18245237

RESUMEN

The Escherichia coli-Helicobacter pylori shuttle vector pHeL2 was modified to introduce the inducible LacI(q)-pTac system of E. coli, in which the promoters were engineered to be under the control of H. pylori RNA polymerase. The amiE gene promoter of H. pylori was taken to constitutively express the LacI(q) repressor. Expression of the reporter gene lacZ was driven by either pTac (pILL2150) or a modified version of the ureI gene promoter in which one or two LacI-binding sites and/or mutated nucleotides between the ribosomal binding site and the ATG start codon (pILL2153 and pILL2157) were introduced. Promoter activity was evaluated by measuring beta-galactosidase activity. pILL2150 is a tightly regulated expression system suitable for the analysis of genes with low-level expression, while pILL2157 is well adapted for the controlled expression of genes encoding recombinant proteins in H. pylori. To exemplify the usefulness of these tools, we constructed conditional mutants of the putative essential pbp1 and ftsI genes encoding penicillin-binding proteins 1 and 3 of H. pylori, respectively. Both genes were cloned into pILL2150 and introduced in the parental H. pylori strain N6. The chromosomally harbored pbp1 and ftsI genes were then inactivated by replacing them with a nonpolar kanamycin cassette. Inactivation was strictly dependent upon addition of isopropyl-beta-d-thiogalactopyranoside. Hence, we were able to construct the first conditional mutants of H. pylori. Finally, we demonstrated that following in vitro methylation of the recombinant plasmids, these could be introduced into a large variety of H. pylori isolates with different genetic backgrounds.


Asunto(s)
Genes Esenciales , Ingeniería Genética , Vectores Genéticos , Helicobacter pylori/genética , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/fisiología , Datos de Secuencia Molecular , Mutagénesis , Regiones Promotoras Genéticas
13.
Nat Commun ; 8(1): 776, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28974686

RESUMEN

Bacterial cell wall biosynthesis is an essential process that requires the coordinated activity of peptidoglycan biosynthesis enzymes within multi-protein complexes involved in cell division (the "divisome") and lateral wall growth (the "elongasome"). MreC is a structural protein that serves as a platform during wall elongation, scaffolding other essential peptidoglycan biosynthesis macromolecules, such as penicillin-binding proteins. Despite the importance of these multi-partite complexes, details of their architecture have remained elusive due to the transitory nature of their interactions. Here, we present the crystal structures of the soluble PBP2:MreC core elongasome complex from Helicobacter pylori, and of uncomplexed PBP2. PBP2 recognizes the two-winged MreC molecule upon opening of its N-terminal region, revealing a hydrophobic zipper that serves as binding platform. The PBP2:MreC interface is essential both for protein recognition in vitro and maintenance of bacterial shape and growth. This work allows visualization as to how peptidoglycan machinery proteins are scaffolded, revealing interaction regions that could be targeted by tailored inhibitors.Bacterial wall biosynthesis is a complex process that requires the coordination of multiple enzymes. Here, the authors structurally characterize the PBP2:MreC complex involved in peptidoglycan elongation and cross-linking, and demonstrate that its disruption leads to loss of H. pylori shape and inability to sustain growth.


Asunto(s)
Proteínas Bacterianas/química , Pared Celular/metabolismo , Helicobacter pylori/genética , Proteínas de Unión a las Penicilinas/química , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Sitios de Unión , Cristalografía por Rayos X , Helicobacter pylori/metabolismo , Modelos Moleculares , Proteínas de Unión a las Penicilinas/genética , Estructura Terciaria de Proteína , Alineación de Secuencia
14.
Microb Drug Resist ; 22(6): 477-86, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27447281

RESUMEN

The human gastric pathogen, Helicobacter pylori, is becoming increasingly resistant to most available antibiotics. Peptidoglycan (PG) metabolism is essential to eubacteria, hence, an excellent target for the development of new therapeutic strategies. However, our knowledge on PG metabolism in H. pylori remains poor. We have further characterized an isogenic mutant of the amiA gene encoding a N-acetylmuramoyl-l-alanyl amidase. The amiA mutant displayed long chains of unseparated cells, an impaired motility despite the presence of intact flagella and a tolerance to amoxicillin. Interestingly, the amiA mutant was impaired in colonizing the mouse stomach suggesting that AmiA is a valid target in H. pylori for the development of new antibiotics. Using reverse phase high-pressure liquid chromatography, we analyzed the PG muropeptide composition and glycan chain length distribution of strain 26695 and its amiA mutant. The analysis showed that H. pylori lacked muropeptides with a degree of cross-linking higher than dimeric muropeptides. The amiA mutant was also characterized by a decrease of muropeptides carrying 1,6-anhydro-N-acetylmuramic acid residues, which represent the ends of the glycan chains. This correlated with an increase of very long glycan strands in the amiA mutant. It is suggested that these longer glycan strands are trademarks of the division site. Taken together, we show that the low redundancy on genes involved in PG maturation supports H. pylori as an actractive alternative model to study PG metabolism and cell shape regulation.


Asunto(s)
Amidohidrolasas/metabolismo , Proteínas Bacterianas/metabolismo , Infecciones por Helicobacter/patología , Helicobacter pylori/enzimología , Peptidoglicano/metabolismo , Amidohidrolasas/genética , Amoxicilina/farmacología , Animales , Antibacterianos/farmacología , Proteínas Bacterianas/genética , División Celular , Expresión Génica , Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/microbiología , Helicobacter pylori/efectos de los fármacos , Helicobacter pylori/genética , Helicobacter pylori/patogenicidad , Ratones , Ratones Endogámicos C57BL , Ácidos Murámicos/química , Ácidos Murámicos/metabolismo , Mutación , Peptidoglicano/química , Estómago/efectos de los fármacos , Estómago/microbiología , Estómago/patología , Virulencia
15.
FEMS Microbiol Lett ; 243(1): 45-50, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15667999

RESUMEN

In order to elucidate the role of polyphosphate kinase (PPK) during the course of an infection by Helicobacter pylori, PPK deficient mutants were constructed using two genetic backgrounds: Hp141v and X47-2AL. The efficiencies of the parental strains and the derivative mutants at colonizing the gastric mucosa of mice were compared. When animals received the Hp141v and the X47-2AL parental strains, 100% of the mice remained colonized for the duration of the 45 days experiment. In contrast, none of the mice that were given the PPK deficient X47-2AL derivative strain had a detectable bacterial load in their gastric mucosa, while the deficient Hp141v derivative strain was detected in 100%, 20% and 40% of the mice at days 3, 15 and 45 post-inoculation (p.i.), respectively. The absence of PPK expression did not impair the in vitro growth of the ppk mutants. However, the reduced ability of the ppk defective mutants to colonize mice was associated with a significant decrease in both motility and in an accumulation of polyP in the bacterial cells. These results are consistent with an essential role of PPK during the initial steps of colonisation of the mouse gastric mucosa and confirm that PPK may act on the virulence of H. pylori partly through an energy dependent mechanism.


Asunto(s)
Mucosa Gástrica/microbiología , Helicobacter pylori/crecimiento & desarrollo , Fosfotransferasas (Aceptor del Grupo Fosfato)/metabolismo , Animales , Femenino , Infecciones por Helicobacter/microbiología , Helicobacter pylori/enzimología , Helicobacter pylori/aislamiento & purificación , Helicobacter pylori/patogenicidad , Ratones , Ratones Endogámicos C57BL , Mutación , Virulencia
16.
Genome Announc ; 1(6)2013 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-24356847

RESUMEN

Helicobacter pylori is a human-specific pathogen that exclusively inhabits the human gastric mucosa. However, occasionally, humans transmit H. pylori to susceptible animal hosts bred in colonies. Here, we report the genome sequence of strain X47-2AL, isolated from a domestic cat and used in anti-H. pylori immunization studies.

17.
Science ; 342(6161): 971-6, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24264990

RESUMEN

Cyclophosphamide is one of several clinically important cancer drugs whose therapeutic efficacy is due in part to their ability to stimulate antitumor immune responses. Studying mouse models, we demonstrate that cyclophosphamide alters the composition of microbiota in the small intestine and induces the translocation of selected species of Gram-positive bacteria into secondary lymphoid organs. There, these bacteria stimulate the generation of a specific subset of "pathogenic" T helper 17 (pT(H)17) cells and memory T(H)1 immune responses. Tumor-bearing mice that were germ-free or that had been treated with antibiotics to kill Gram-positive bacteria showed a reduction in pT(H)17 responses, and their tumors were resistant to cyclophosphamide. Adoptive transfer of pT(H)17 cells partially restored the antitumor efficacy of cyclophosphamide. These results suggest that the gut microbiota help shape the anticancer immune response.


Asunto(s)
Antineoplásicos/uso terapéutico , Traslocación Bacteriana/efectos de los fármacos , Ciclofosfamida/uso terapéutico , Inmunosupresores/uso terapéutico , Intestino Delgado/microbiología , Microbiota/fisiología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Traslado Adoptivo , Animales , Antibacterianos/administración & dosificación , Vida Libre de Gérmenes , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/fisiología , Memoria Inmunológica , Tejido Linfoide/inmunología , Tejido Linfoide/microbiología , Ratones , Microbiota/efectos de los fármacos , Células Th17/inmunología , Células Th17/trasplante
18.
Antioxid Redox Signal ; 17(9): 1190-200, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22236381

RESUMEN

AIMS: The ability of pathogens to cope with the damaging effects of nitric oxide (NO), present in certain host niches and produced by phagocytes that support innate immunity, relies on multiple strategies that include the action of detoxifying enzymes. As for many other pathogens, these systems remained unknown for Helicobacter pylori. This work aimed at identifying and functionally characterizing an H. pylori system involved in NO protection. RESULTS: In the present work, the hp0013 gene of H. pylori is shown to be related to NO resistance, as its inactivation increases the susceptibility of H. pylori to nitrosative stress, and significantly decreases the NADPH-dependent NO reduction activity of H. pylori cells. The recombinant HP0013 protein is able to complement an NO reductase-deficient Escherichia coli strain and exhibits significant NO reductase activity. Mutation of hp0013 renders H. pylori more vulnerable to nitric oxide synthase-dependent macrophage killing, and decreases the ability of the pathogen to colonize mice stomachs. INNOVATION: Phylogenetic studies reveal that HP0013, which shares no significant amino acid sequence similarity to the other so far known microbial NO detoxifiers, belongs to a novel family of proteins with a widespread distribution in the microbial world. CONCLUSION: H. pylori HP0013 represents an unprecedented enzymatic NO detoxifying system for the in vivo microbial protection against nitrosative stress.


Asunto(s)
Proteínas Bacterianas/metabolismo , Helicobacter pylori/metabolismo , Óxido Nítrico/metabolismo , Proteínas Bacterianas/clasificación , Proteínas Bacterianas/genética , Helicobacter pylori/enzimología , Helicobacter pylori/genética , Filogenia , Especies de Nitrógeno Reactivo/metabolismo
19.
mBio ; 1(3)2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-21151756

RESUMEN

Despite recent advances in our understanding of how Helicobacter pylori causes disease, the factors that allow this pathogen to persist in the stomach have not yet been fully characterized. To identify new virulence factors in H. pylori, we generated low-infectivity variants of a mouse-colonizing H. pylori strain using the classical technique of in vitro attenuation. The resulting variants and their highly infectious progenitor bacteria were then analyzed by global gene expression profiling. The gene expression levels of five open reading frames (ORFs) were significantly reduced in low-infectivity variants, with the most significant changes observed for ORFs HP1583 and HP1582. These ORFs were annotated as encoding homologs of the Escherichia coli vitamin B(6) biosynthesis enzymes PdxA and PdxJ. Functional complementation studies with E. coli confirmed H. pylori PdxA and PdxJ to be bona fide homologs of vitamin B(6) biosynthesis enzymes. Importantly, H. pylori PdxA was required for optimal growth in vitro and was shown to be essential for chronic colonization in mice. In addition to having a well-known metabolic role, vitamin B(6) is necessary for the synthesis of glycosylated flagella and for flagellum-based motility in H. pylori. Thus, for the first time, we identify vitamin B(6) biosynthesis enzymes as novel virulence factors in bacteria. Interestingly, pdxA and pdxJ orthologs are present in a number of human pathogens, but not in mammalian cells. We therefore propose that PdxA/J enzymes may represent ideal candidates for therapeutic targets against bacterial pathogens.


Asunto(s)
Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Helicobacter pylori/patogenicidad , Vitamina B 6/biosíntesis , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Femenino , Mucosa Gástrica/microbiología , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Virulencia
20.
EMBO J ; 26(1): 232-41, 2007 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-17159901

RESUMEN

To date, the biological role of prokaryotic K(+) channels remains unknown. Helicobacter pylori contains a gene encoding a putative K(+) channel (HpKchA) of the two-transmembrane RCK (regulation of K(+) conductance) domain family, but lacks known bacterial K(+) uptake systems. A H. pylori DeltahpKchA mutant presented a strong growth defect at low K(+) concentration, which was compensated by KCl addition. The role of the separate RCK domain was investigated in H. pylori by mutagenesis of its internal start codon, which led to a K(+)-dependent intermediate growth phenotype, consistent with RCK activating channel function. Tagging HpKchA C-terminally, we detected a 1:1 stoichiometry of the full-length HpKchA and the separate RCK domain. We constructed single amino-acid exchanges within the unusual selectivity filter of HpKchA (ATGFGA) in H. pylori and observed complete loss (G74A), a slight defect (G76A or F75G) or wild-type (A77D) channel function. HpKchA was essential for colonization of the murine stomach. These data show, for the first time, a biological function for a prokaryotic K(+) channel, as a K(+) uptake system, essential for the persistence of H. pylori in the gastric environment.


Asunto(s)
Mucosa Gástrica/microbiología , Helicobacter pylori/metabolismo , Canales de Potasio/química , Potasio/farmacocinética , Secuencia de Aminoácidos , Animales , Codón Iniciador , Citoplasma/metabolismo , Concentración de Iones de Hidrógeno , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Potasio/química , Canales de Potasio/fisiología , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda