Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Open Biol ; 4(10)2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25274119

RESUMEN

The non-integrin laminin receptor (LAMR1/RPSA) and galectin-3 (Gal-3) are multi-functional host molecules with roles in diverse pathological processes, particularly of infectious or oncogenic origins. Using bimolecular fluorescence complementation and confocal imaging, we demonstrate that the two proteins homo- and heterodimerize, and that each isotype forms a distinct cell surface population. We present evidence that the 37 kDa form of LAMR1 (37LRP) is the precursor of the previously described 67 kDa laminin receptor (67LR), whereas the heterodimer represents an entity that is distinct from this molecule. Site-directed mutagenesis confirmed that the single cysteine (C(173)) of Gal-3 or lysine (K(166)) of LAMR1 are critical for heterodimerization. Recombinant Gal-3, expressed in normally Gal-3-deficient N2a cells, dimerized with endogenous LAMR1 and led to a significantly increased number of internalized bacteria (Neisseria meningitidis), confirming the role of Gal-3 in bacterial invasion. Contact-dependent cross-linking determined that, in common with LAMR1, Gal-3 binds the meningococcal secretin PilQ, in addition to the major pilin PilE. This study adds significant new mechanistic insights into the bacterial-host cell interaction by clarifying the nature, role and bacterial ligands of LAMR1 and Gal-3 isotypes during colonization.


Asunto(s)
Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Galectina 3/metabolismo , Regulación de la Expresión Génica , Neisseria meningitidis/metabolismo , Receptores de Laminina/metabolismo , Animales , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Reactivos de Enlaces Cruzados/química , Humanos , Enlace de Hidrógeno , Integrinas/metabolismo , Lactosa/química , Ligandos , Ratones , Microscopía Confocal , Microscopía Fluorescente , Modelos Moleculares , Conformación Molecular , Mutagénesis Sitio-Dirigida , Multimerización de Proteína
2.
Open Biol ; 4: 130202, 2014 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-24451549

RESUMEN

Campylobacter jejuni is an important cause of human foodborne gastroenteritis; strategies to prevent infection are hampered by a poor understanding of the complex interactions between host and pathogen. Previous work showed that C. jejuni could bind human histo-blood group antigens (BgAgs) in vitro and that BgAgs could inhibit the binding of C. jejuni to human intestinal mucosa ex vivo. Here, the major flagella subunit protein (FlaA) and the major outer membrane protein (MOMP) were identified as BgAg-binding adhesins in C. jejuni NCTC11168. Significantly, the MOMP was shown to be O-glycosylated at Thr(268); previously only flagellin proteins were known to be O-glycosylated in C. jejuni. Substitution of MOMP Thr(268) led to significantly reduced binding to BgAgs. The O-glycan moiety was characterized as Gal(ß1-3)-GalNAc(ß1-4)-GalNAc(ß1-4)-GalNAcα1-Thr(268); modelling suggested that O-glycosylation has a notable effect on the conformation of MOMP and this modulates BgAg-binding capacity. Glycosylation of MOMP at Thr(268) promoted cell-to-cell binding, biofilm formation and adhesion to Caco-2 cells, and was required for the optimal colonization of chickens by C. jejuni, confirming the significance of this O-glycosylation in pathogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Antígenos de Grupos Sanguíneos/metabolismo , Campylobacter jejuni/metabolismo , Polisacáridos/metabolismo , Porinas/metabolismo , Animales , Proteínas Bacterianas/química , Sitios de Unión , Biopelículas , Antígenos de Grupos Sanguíneos/química , Células CACO-2 , Pollos , Flagelina/química , Flagelina/genética , Flagelina/metabolismo , Glicosilación , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Mutagénesis , Polisacáridos/química , Porinas/química , Unión Proteica , Estructura Terciaria de Proteína
3.
Open Biol ; 3(10): 130048, 2013 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-24107297

RESUMEN

Interactions between commensal pathogens and hosts are critical for disease development but the underlying mechanisms for switching between the commensal and virulent states are unknown. We show that the human pathogen Neisseria meningitidis, the leading cause of pyogenic meningitis, can modulate gene expression via uptake of host pro-inflammatory cytokines leading to increased virulence. This uptake is mediated by type IV pili (Tfp) and reliant on the PilT ATPase activity. Two Tfp subunits, PilE and PilQ, are identified as the ligands for TNF-α and IL-8 in a glycan-dependent manner, and their deletion results in decreased virulence and increased survival in a mouse model. We propose a novel mechanism by which pathogens use the twitching motility mode of the Tfp machinery for sensing and importing host elicitors, aligning with the inflamed environment and switching to the virulent state.


Asunto(s)
Citocinas/metabolismo , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/metabolismo , Interleucina-8/metabolismo , Meningitis Bacterianas/microbiología , Neisseria meningitidis/patogenicidad , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Proteínas Fimbrias/genética , Fimbrias Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Humanos , Ligandos , Meningitis Bacterianas/metabolismo , Ratones , Ratones Transgénicos , Neisseria meningitidis/genética , Neisseria meningitidis/metabolismo , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
4.
Infect Immun ; 78(10): 4261-7, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20643857

RESUMEN

The genomes of Helicobacter species colonizing the mammalian gastric mucosa (like Helicobacter pylori) contain a large number of genes annotated as iron acquisition genes but only few nickel acquisition genes, which contrasts with the central position of nickel in the urease-mediated acid resistance of these gastric pathogens. In this study we have investigated the predicted iron and nickel acquisition systems of the ferret pathogen Helicobacter mustelae. The expression of the outer membrane protein-encoding frpB2 gene was iron and Fur repressed, whereas the expression of the ABC transporter genes fecD and ceuE was iron and Fur independent. The inactivation of the two tonB genes showed that TonB1 is required for heme utilization, whereas the absence of TonB2 only marginally affected iron-dependent growth but led to reduced cellular nickel content and urease activity. The inactivation of the fecD and ceuE ABC transporter genes did not affect iron levels but resulted in significantly reduced urease activity and cellular nickel content. Surprisingly, the inactivation of the nixA nickel transporter gene affected cellular nickel content and urease activity only when combined with the inactivation of other nickel acquisition genes, like fecD or ceuE. The FecDE ABC transporter is not specific for nickel, since an fecD mutant also showed reduced cellular cobalt levels and increased cobalt resistance. We conclude that the H. mustelae fecDE and ceuE genes encode an ABC transporter involved in nickel and cobalt acquisition, which works independently of the nickel transporter NixA, while TonB2 is required primarily for nickel acquisition, with TonB1 being required for heme utilization.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Bacterianas/metabolismo , Cobalto/metabolismo , Helicobacter mustelae/metabolismo , Proteínas de la Membrana/metabolismo , Níquel/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Animales , Proteínas Bacterianas/clasificación , Proteínas Bacterianas/genética , Transporte Biológico , Hurones , Regulación Bacteriana de la Expresión Génica/fisiología , Genoma Bacteriano , Helicobacter mustelae/genética , Proteínas de la Membrana/clasificación , Proteínas de la Membrana/genética , Mutación , Ureasa/metabolismo
5.
Biometals ; 23(1): 145-59, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19894125

RESUMEN

The NikR protein is a nickel-responsive regulator, which in the gastric pathogen Helicobacter pylori controls expression of nickel-transporters and the nickel-cofactored urease acid resistance determinant. Although NikR-DNA interaction has been well studied, the Helicobacter NikR operator site remains poorly defined. In this study we have identified the NikR operators in the promoters of two inversely nickel-regulated urease operons (ureAB and ureA2B2) in the ferret pathogen Helicobacter mustelae, and have used bioinformatic approaches for the prediction of putative NikR operators in the genomes of four urease-positive Helicobacter species. Helicobacter mustelae NikR bound to the ureA2 promoter to a sequence overlapping with the -35 promoter region, leading to repression. In contrast, NikR binding to a site far upstream of the canonical sigma(80) promoter in the H. mustelae ureA promoter resulted in transcriptional induction, similar to the situation in H. pylori. Using H. pylori NikR operators and the newly identified H. mustelae NikR operators a new consensus sequence was generated (TRWYA-N(15)-TRWYA), which was used to screen the genomes of four urease-positive Helicobacter species (H. mustelae, H. pylori, H. acinonychis and H. hepaticus) for putative NikR-regulated promoters. One of these novel putative NikR-regulated promoters in H. mustelae is located upstream of a putative TonB-dependent outer membrane protein designated NikH, which displayed nickel-responsive expression. Insertional inactivation of the nikH gene in H. mustelae resulted in a significant decrease in urease activity, and this phenotype was complemented by nickel-supplementation of the growth medium, suggesting a function for NikH in nickel transport across the outer membrane. In conclusion, the H. mustelae NikR regulator directly controls nickel-responsive regulation of ureases and metal transporters. The improved consensus NikR operator sequence allows the prediction of additional NikR targets in Helicobacter genomes, as demonstrated by the identification of a new nickel-repressed outer membrane protein in H. mustelae.


Asunto(s)
Proteínas de Transporte de Catión/genética , Helicobacter mustelae/enzimología , Helicobacter mustelae/genética , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Ureasa/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Helicobacter mustelae/metabolismo , Proteínas Represoras/química , Ureasa/metabolismo
6.
Helicobacter ; 14(1): 72-9, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19191900

RESUMEN

BACKGROUND: Gastrointestinal infections with pathogenic Helicobacter species are commonly treated with combination therapies, which often include amoxicillin. Although this treatment is effective for eradication of Helicobacter pylori, the few existing reports are less clear about antibiotic susceptibility of other Helicobacter species. In this study we have determined the susceptibility of gastric and enterohepatic Helicobacter species to amoxicillin, and have investigated the mechanism of amoxicillin resistance in Helicobacter hepaticus. MATERIALS AND METHODS: The minimal inhibitory concentration (MIC) of antimicrobial compounds was determined by E-test and agar/broth dilution assays. The hefA gene of H. hepaticus was inactivated by insertion of a chloramphenicol resistance gene. Transcription was measured by quantitative real-time polymerase chain reaction. RESULTS: Three gastric Helicobacter species (H. pylori, H. mustelae, and H. acinonychis) were susceptible to amoxicillin (MIC < 0.25 mg/L). In contrast, three enterohepatic Helicobacter species (H. rappini, H. bilis, and H. hepaticus) were resistant to amoxicillin (MIC of 8, 16, and 6-64 mg/L, respectively). There was no detectable beta-lactamase activity in H. hepaticus, and inhibition of beta-lactamases did not change the MIC of amoxicillin of H. hepaticus. A H. hepaticus hefA (hh0224) mutant, encoding a TolC-component of a putative efflux system, resulted in loss of amoxicillin resistance (MIC 0.25 mg/L), and also resulted in increased sensitivity to bile acids. Finally, transcription of the hefA gene was not responsive to amoxicillin, but induced by bile acids. CONCLUSIONS: Rodents are frequently colonized by a variety of enterohepatic Helicobacter species, and this may affect their global health status and intestinal inflammatory responses. Animal facilities should have treatment strategies for Helicobacter infections, and hence resistance of enterohepatic Helicobacter species to amoxicillin should be considered when designing eradication programs.


Asunto(s)
Amoxicilina/farmacología , Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Helicobacter hepaticus/genética , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Animales , Regulación Bacteriana de la Expresión Génica , Helicobacter hepaticus/efectos de los fármacos , Helicobacter hepaticus/metabolismo , Humanos , Pruebas de Sensibilidad Microbiana , Mutación
7.
Environ Microbiol ; 10(10): 2586-97, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18564183

RESUMEN

The acidic gastric environment of mammals can be chronically colonized by pathogenic Helicobacter species, which use the nickel-dependent urea-degrading enzyme urease to confer acid resistance. Nickel availability in the mammal host is low, being mostly restricted to vegetarian dietary sources, and thus Helicobacter species colonizing carnivores may be subjected to episodes of nickel deficiency and associated acid sensitivity. The aim of this study was to investigate how these Helicobacter species have adapted to the nickel-restricted diet of their carnivorous host. Three carnivore-colonizing Helicobacter species express a second functional urea-degrading urease enzyme (UreA2B2), which functions as adaptation to nickel deficiency. UreA2B2 was not detected in seven other Helicobacter species, and is in Helicobacter mustelae only expressed in nickel-restricted conditions, and its expression was higher in iron-rich conditions. In contrast to the standard urease UreAB, UreA2B2 does not require activation by urease or hydrogenase accessory proteins, which mediate nickel incorporation into these enzymes. Activity of either UreAB or UreA2B2 urease allowed survival of a severe acid shock in the presence of urea, demonstrating a functional role for UreA2B2 in acid resistance. Pathogens often express colonization factors which are adapted to their host. The UreA2B2 urease could represent an example of pathogen adaptation to the specifics of the diet of their carnivorous host, rather than to the host itself.


Asunto(s)
Helicobacter mustelae/enzimología , Níquel/metabolismo , Ureasa/biosíntesis , Ácidos/farmacología , Animales , Antibacterianos/farmacología , Inducción Enzimática , Perfilación de la Expresión Génica , Orden Génico , Helicobacter mustelae/efectos de los fármacos , Viabilidad Microbiana , Operón
8.
FEMS Immunol Med Microbiol ; 50(2): 273-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17298583

RESUMEN

Urease activity is vital for gastric colonization by Helicobacter species, such as the animal pathogen Helicobacter felis. Here it is demonstrated that H. felis expresses two independent, and distinct urease systems. H. felis isolate CS1 expressed two proteins of 67 and 70 kDa reacting with antibodies to H. pylori urease. The 67-kDa protein was identified as the UreB urease subunit, whereas the N-terminal amino acid sequence of the 70-kDa protein displayed 58% identity with the UreB protein and was tentatively named UreB2. The gene encoding the UreB2 protein was identified and located in a gene cluster named ureA2B2. Inactivation of ureB led to complete absence of urease activity, whereas inactivation of ureB2 resulted in decreased urease activity. Although the exact function of the UreA2B2 system is still unknown, it is conceivable that UreA2B2 may contribute to pathogenesis of H. felis infection through a yet unknown mechanism.


Asunto(s)
Helicobacter felis/enzimología , Helicobacter felis/genética , Ureasa/genética , Anticuerpos Antibacterianos/metabolismo , Proteínas Bacterianas/genética , Eliminación de Gen , Datos de Secuencia Molecular , Peso Molecular , Familia de Multigenes , Mutagénesis Insercional , Filogenia , Análisis de Secuencia de ADN , Análisis de Secuencia de Proteína , Homología de Secuencia de Aminoácido , Ureasa/análisis , Ureasa/metabolismo , Factores de Virulencia/genética
9.
Biometals ; 20(3-4): 417-29, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17294126

RESUMEN

Helicobacter species are among the most successful colonizers of the mammalian gastrointestinal and hepatobiliary tract. Colonization is usually lifelong, indicating that Helicobacter species have evolved intricate mechanisms of dealing with stresses encountered during colonization of host tissues, like restriction of essential metal ions. The recent availability of genome sequences of the human gastric pathogen Helicobacter pylori, the murine enterohepatic pathogen Helicobacter hepaticus and the unannotated genome sequence of the ferret gastric pathogen Helicobacter mustelae has allowed for comparative genome analyses. In this review we present such analyses for metal transporters, metal-storage and metal-responsive regulators in these three Helicobacter species, and discuss possible contributions of the differences in metal metabolism in adaptation to the gastric or enterohepatic niches occupied by Helicobacter species.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Helicobacter , Metales/metabolismo , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Transporte Biológico/fisiología , Helicobacter/genética , Helicobacter/metabolismo , Homeostasis , Humanos , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Datos de Secuencia Molecular
10.
Infect Immun ; 74(12): 6821-8, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17015456

RESUMEN

The transition metal nickel plays an important role in gastric colonization and persistence of the important human pathogen Helicobacter pylori, as it is the cofactor of the abundantly produced acid resistance factor urease. Nickel uptake through the inner membrane is mediated by the NixA protein, and the expression of NixA is controlled by the NikR regulatory protein. Here we report that NikR also controls the nickel-responsive expression of the FecA3 (HP1400) and FrpB4 (HP1512) outer membrane proteins (OMPs), as well as the nickel-responsive expression of an ExbB-ExbD-TonB system, which may function in energization of outer membrane transport. Transcription and expression of the frpB4 and fecA3 genes were repressed by nickel in wild-type H. pylori 26695, but they were independent of nickel and derepressed in an isogenic nikR mutant. Both the frpB4 and fecA3 genes were transcribed from a promoter directly upstream of their start codon. Regulation by NikR was mediated via nickel-dependent binding to specific operators overlapping either the +1 or -10 sequence in the frpB4 and fecA3 promoters, respectively, and these operators contained sequences resembling the proposed H. pylori NikR recognition sequence (TATWATT-N(11)-AATWATA). Transcription of the HP1339-1340-1341 operon encoding the ExbB2-ExbD2-TonB2 complex was also regulated by nickel and NikR, but not by Fur and iron. In conclusion, H. pylori NikR controls nickel-responsive expression of the HP1400 (FecA3) and HP1512 (FrpB4) OMPs. We hypothesize that these two NikR-regulated OMPs may participate in the uptake of complexed nickel ions and that this process is energized by the NikR-regulated ExbB2-ExbD2-TonB2 system, another example of the specific adaptation of H. pylori to the gastric lifestyle.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/genética , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/genética , Níquel/metabolismo , Proteínas Represoras/fisiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/fisiología , Secuencia de Bases , Regulación hacia Abajo , Helicobacter pylori/efectos de los fármacos , Hierro/metabolismo , Hierro/farmacología , Datos de Secuencia Molecular , Níquel/farmacología , Operón/efectos de los fármacos , Operón/genética , Regiones Promotoras Genéticas , Proteínas Represoras/genética , Transcripción Genética/efectos de los fármacos
11.
Helicobacter ; 11(3): 181-7, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16684266

RESUMEN

BACKGROUND: Amoxicillin-based therapies are highly effective for the treatment of Helicobacter pylori infections, but the efficacy may decrease as the incidence of amoxicillin resistance is increasing. So far, the molecular mechanism underlying stable amoxicillin resistance has only been identified for a few naturally occurring amoxicillin-resistant (Amx) H. pylori isolates, and is mediated by mutations in penicillin-binding protein 1A (PBP1A). In this study the molecular mechanism underlying amoxicillin resistance of seven additional Amx H. pylori isolates has been established. METHODS: H. pylori strain 26695 (minimal inhibitory concentration (MIC) 0.125 mg/l) was naturally transformed with total DNA and pbp1A polymerase chain reaction (PCR) products from the seven Amx H. pylori isolates, and the MIC of amoxicillin and pbp1A gene sequence of the obtained Amx transformants were determined. RESULTS: Replacement of the wild-type pbp1A gene of H. pylori reference strain 26695 by the pbp1A gene of the Amx H. pylori isolates resulted in an increased MIC (0.5-1.0 mg/l). Sequence analysis of the smallest PBP1A fragments able to transfer the resistance indicated that several amino acid substitutions in or adjacent to the second (SKN402-404) and third (KTG555-557) conserved penicillin-binding protein motifs (PBP-motifs) mediate amoxicillin resistance in H. pylori. This was confirmed by site-directed mutagenesis using oligonucleotides that contained defined mutations in or adjacent to these PBP-motifs. CONCLUSION: In naturally occurring Amx H. pylori isolates, amoxicillin resistance is mediated by various mutational changes located in or adjacent to the second and third PBP-motifs of the PBP1A. Although we cannot exclude the role of the other genes in amoxicillin resistance, it is likely that multiple mutational changes in the PBP1A gene are the predominant cause of amoxicillin resistance in H. pylori. The findings of this study currently preclude the rapid detection of amoxicillin resistance in H. pylori by molecular tests.


Asunto(s)
Amoxicilina/farmacología , Antibacterianos/farmacología , Helicobacter pylori/efectos de los fármacos , Mutación , Resistencia a las Penicilinas , Proteínas de Unión a las Penicilinas/genética , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Humanos , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Resistencia a las Penicilinas/genética , Proteínas de Unión a las Penicilinas/química , Análisis de Secuencia de ADN
12.
Microbiology (Reading) ; 151(Pt 12): 3989-3995, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16339943

RESUMEN

Helicobacter hepaticus is a pathogen of rodents, which causes diverse enteric and hepatic inflammatory diseases and malignancies. The urease enzyme is an important colonization factor of gastric Helicobacter species like Helicobacter pylori, but little is known about the role and regulation of urease in enterohepatic Helicobacter species. Here it is reported that urease activity of H. hepaticus does not contribute to acid resistance, and that it is nickel-responsive at the post-translational level. H. hepaticus strain ATCC 51449 did not grow or survive at pH 3.0, and supplementation with urea or NiCl2 did not abrogate this acid sensitivity. Furthermore, urease enzyme activity of H. hepaticus was acid-independent, which contrasts with the acid-induced urease system of H. pylori. Nickel supplementation of Brucella medium resulted in a tenfold increase in urease activity in both H. hepaticus and H. pylori, but the maximum level of urease activity in H. hepaticus was still three- to fivefold lower when compared to H. pylori in the same conditions. The increase in urease activity of H. hepaticus was not associated with elevation of urease mRNA or protein levels. Inhibition of protein synthesis by chloramphenicol did not affect nickel-responsive induction of urease activity in H. hepaticus, and confirmed that nickel induction occurs at the post-translational level, probably by activation of preformed apo-enzyme. In conclusion, both the role of the urease enzyme and the regulation of urease activity differ between the enterohepatic pathogen H. hepaticus and the gastric pathogen H. pylori.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Helicobacter hepaticus/enzimología , Helicobacter pylori/enzimología , Níquel/farmacología , Ureasa/metabolismo , Helicobacter hepaticus/genética , Helicobacter hepaticus/crecimiento & desarrollo , Helicobacter pylori/genética , Helicobacter pylori/crecimiento & desarrollo , Ureasa/genética
13.
Infect Immun ; 73(11): 7252-8, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16239520

RESUMEN

The NikR protein is a nickel-dependent regulatory protein which is a member of the ribbon-helix-helix family of transcriptional regulators. The gastric pathogen Helicobacter pylori expresses a NikR ortholog, which was previously shown to mediate regulation of metal metabolism and urease expression, but the mechanism governing the diverse regulatory effects had not been described until now. In this study it is demonstrated that NikR can regulate H. pylori nickel metabolism by directly controlling transcriptional repression of NixA-mediated nickel uptake and transcriptional induction of urease expression. Mutation of the nickel uptake gene nixA in an H. pylori 26695 nikR mutant restored the ability to grow in Brucella media supplemented with 200 microM NiCl2 but did not restore nickel-dependent induction of urease expression. Nickel-dependent binding of NikR to the promoter of the nixA gene resulted in nickel-repressed transcription, whereas nickel-dependent binding of NikR to the promoter of the ureA gene resulted in nickel-induced transcription. Subsequent analysis of NikR binding to the nixA and ureA promoters showed that the regulatory effect was dependent on the location of the NikR-recognized binding sequence. NikR recognized the region from -13 to +21 of the nixA promoter, encompassing the +1 and -10 region, and this binding resulted in repression of nixA transcription. In contrast, NikR bound to the region from -56 to -91 upstream of the ureA promoter, resulting in induction of urease transcription. In conclusion, the NikR protein is able to function both as a repressor and as an activator of gene transcription, depending on the position of the binding site.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Helicobacter pylori/genética , Níquel/farmacología , Proteínas Represoras/metabolismo , Transcripción Genética/efectos de los fármacos , Proteínas Bacterianas/genética , Secuencia de Bases , Proteínas de Transporte de Catión/genética , Regulación Bacteriana de la Expresión Génica/genética , Helicobacter pylori/efectos de los fármacos , Helicobacter pylori/enzimología , Datos de Secuencia Molecular , Regiones Operadoras Genéticas/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Transcripción Genética/genética , Ureasa/genética
14.
J Bacteriol ; 187(11): 3687-92, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15901691

RESUMEN

Maintaining iron homeostasis is a necessity for all living organisms, as free iron augments the generation of reactive oxygen species like superoxide anions, at the risk of subsequent lethal cellular damage. The iron-responsive regulator Fur controls iron metabolism in many bacteria, including the important human pathogen Helicobacter pylori, and thus is directly or indirectly involved in regulation of oxidative stress defense. Here we demonstrate that Fur is a direct regulator of the H. pylori iron-cofactored superoxide dismutase SodB, which is essential for the defense against toxic superoxide radicals. Transcription of the sodB gene was iron induced in H. pylori wild-type strain 26695, resulting in expression of the SodB protein in iron-replete conditions but an absence of expression in iron-restricted conditions. Mutation of the fur gene resulted in constitutive, iron-independent expression of SodB. Recombinant H. pylori Fur protein bound with low affinity to the sodB promoter region, but addition of the iron substitute Mn2+ abolished binding. The operator sequence of the iron-free form of Fur, as identified by DNase I footprinting, was located directly upstream of the sodB gene at positions -5 to -47 from the transcription start site. The direct role of Fur in regulation of the H. pylori sodB gene contrasts with the small-RNA-mediated sodB regulation observed in Escherichia coli. In conclusion, H. pylori Fur is a versatile regulator involved in many pathways essential for gastric colonization, including superoxide stress defense.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Helicobacter pylori/genética , Hierro/metabolismo , Estrés Oxidativo/fisiología , Proteínas Represoras/metabolismo , Superóxido Dismutasa/genética , Secuencia de Bases , Huella de ADN , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/metabolismo , Regiones Promotoras Genéticas/fisiología , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Transcripción Genética
15.
Microbiology (Reading) ; 151(Pt 2): 533-546, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15699202

RESUMEN

Intracellular iron homeostasis is a necessity for almost all living organisms, since both iron restriction and iron overload can result in cell death. The ferric uptake regulator protein, Fur, controls iron homeostasis in most Gram-negative bacteria. In the human gastric pathogen Helicobacter pylori, Fur is thought to have acquired extra functions to compensate for the relative paucity of regulatory genes. To identify H. pylori genes regulated by iron and Fur, we used DNA array-based transcriptional profiling with RNA isolated from H. pylori 26695 wild-type and fur mutant cells grown in iron-restricted and iron-replete conditions. Sixteen genes encoding proteins involved in metal metabolism, nitrogen metabolism, motility, cell wall synthesis and cofactor synthesis displayed iron-dependent Fur-repressed expression. Conversely, 16 genes encoding proteins involved in iron storage, respiration, energy metabolism, chemotaxis, and oxygen scavenging displayed iron-induced Fur-dependent expression. Several Fur-regulated genes have been previously shown to be essential for acid resistance or gastric colonization in animal models, such as those encoding the hydrogenase and superoxide dismutase enzymes. Overall, there was a partial overlap between the sets of genes regulated by Fur and those previously identified as growth-phase, iron or acid regulated. Regulatory patterns were confirmed for five selected genes using Northern hybridization. In conclusion, H. pylori Fur is a versatile regulator involved in many pathways essential for gastric colonization. These findings further delineate the central role of Fur in regulating the unique capacity of H. pylori to colonize the human stomach.


Asunto(s)
Proteínas Bacterianas/metabolismo , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/metabolismo , Hierro/metabolismo , Proteínas Represoras/metabolismo , Proteínas Bacterianas/genética , Secuencia de Bases , Helicobacter pylori/genética , Helicobacter pylori/crecimiento & desarrollo , Humanos , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteoma , Proteínas Represoras/genética , Transcripción Genética
16.
FEMS Immunol Med Microbiol ; 41(2): 161-7, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15145461

RESUMEN

Colonization with Helicobacter pylori always results in chronic gastritis, which is controlled by infiltration of mononuclear cells and the subsequent release of cytokines like interleukin (IL)-12. To identify H. pylori factors involved in inducing cytokine production in mononuclear cells, a random H. pylori mutant library was screened for the inability to induce IL-12 production in monocyte THP-1 cells. Of the 231 random mutants screened, one mutant (M1) showed a consistent twofold decrease in the amount of IL-12 induction compared to the parental strain 1061 (P <0.01). Further characterization of mutant M1 revealed that the kanamycin resistance cassette had integrated in the jhp0945 gene, which is situated in an H. pylori strain-specific plasticity region. Three reference strains possessing this plasticity region induced significantly higher amounts of IL-12 when compared to the H. pylori 26695 reference strain, which does not possess this plasticity region. The role in disease outcome of jhp0945 as well as the neighbouring plasticity region genes jhp0947 and jhp049 was assessed in a Dutch population cohort. Firstly, the presence of jhp0947 was completely linked with that of jhp0949 and was roughly associated with jhp0945 (P=0.072), but not with the cag pathogenicity island (PAI) (P=0.464). The presence of the jhp0947 and jhp0949 genes, but not of jhp0945, was significantly associated with duodenal ulcer disease when compared to gastritis (P=0.027). Therefore, the jhp0947-jhp0949 locus may be a novel putative H. pylori marker for disease outcome independent of the cag PAI.


Asunto(s)
Úlcera Duodenal/microbiología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Interleucina-12/inmunología , Monocitos/inmunología , Monocitos/microbiología , Células Cultivadas , Estudios de Cohortes , ADN Bacteriano/química , ADN Bacteriano/genética , Úlcera Duodenal/inmunología , Variación Genética , Infecciones por Helicobacter/sangre , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Humanos , Interleucina-12/genética , Resistencia a la Kanamicina/genética , Modelos Logísticos , Monocitos/metabolismo , Mutagénesis Insercional , Países Bajos , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN
17.
Infect Immun ; 72(2): 766-73, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14742519

RESUMEN

Although the adaptive mechanisms allowing the gastric pathogen Helicobacter pylori to survive acid shocks have been well documented, the mechanisms allowing growth at mildly acidic conditions (pH approximately 5.5) are still poorly understood. Here we demonstrate that H. pylori strain 26695 increases the transcription and activity of its urease, amidase, and formamidase enzymes four- to ninefold in response to growth at pH 5.5. Supplementation of growth medium with NiCl2 resulted in a similar induction of urease activity (at low NiCl2 concentration) and amidase activity (at > or = 500 micro M NiCl2) but did not affect formamidase activity. Mutation of the fur gene, which encodes an iron-responsive repressor of both amidases, resulted in a constitutively high level of amidase and formamidase activity at either pH but did not affect urease activity at pH 7.0 or pH 5.5. In contrast, mutation of the nikR gene, encoding the nickel-responsive activator of urease expression, resulted in a significant reduction of acid-responsive induction of amidase and formamidase activity. Finally, acid-responsive repression of fur transcription was absent in the H. pylori nikR mutant, whereas transcription of the nikR gene itself was increased at pH 5.5 in wild-type H. pylori. We hypothesize that H. pylori uses a repressor cascade to respond to low pH, with NikR initiating the response directly via the urease operon and indirectly via the members of the Fur regulon.


Asunto(s)
Amoníaco/metabolismo , Proteínas de Escherichia coli , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/metabolismo , Níquel/farmacología , Proteínas Represoras/fisiología , Ácidos , Proteínas Bacterianas/fisiología , Medios de Cultivo , Helicobacter pylori/genética , Concentración de Iones de Hidrógeno , Transcripción Genética , Activación Transcripcional , Ureasa/fisiología
18.
FEMS Immunol Med Microbiol ; 38(1): 45-51, 2003 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-12900054

RESUMEN

The transcriptional regulation, genetic variation and clinical relevance of the strain-specific hsp12 gene of the human gastric pathogen Helicobacter pylori were investigated. Although the transcription of the hsp12 gene in H. pylori strain 1061 was induced by growth under iron-, pH- and temperature-stress conditions, the gene was not essential for growth under these stress conditions. The locus containing the hsp12 gene showed considerable genetic variation. A total of eight different strain-specific alleles were identified, of which three are mosaic variants of the hsp12 gene and five that are unrelated to the hsp12 gene. The hsp12 locus of six paired sets of strains obtained from patients with 7-10-year time intervals remained unaltered, indicating that genetic variation does not occur during chronic infection. No significant association was found between the presence of a hsp12 gene and peptic ulcer disease in clinical isolates obtained from 26 patients. The stress-regulated, strain-specific hsp12 genes may be involved in adaptation of individual H. pylori strains to their specific hosts, and contribute to long-term colonization of the gastric niche.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Proteínas de Choque Térmico/genética , Helicobacter pylori/genética , Úlcera Duodenal/genética , Úlcera Duodenal/microbiología , Variación Genética , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Helicobacter pylori/clasificación , Helicobacter pylori/metabolismo , Humanos , Datos de Secuencia Molecular , Proteínas de Saccharomyces cerevisiae
19.
J Biol Chem ; 278(11): 9052-7, 2003 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-12499381

RESUMEN

The production of high levels of ammonia allows the human gastric pathogen Helicobacter pylori to survive the acidic conditions in the human stomach. H. pylori produces ammonia through urease-mediated degradation of urea, but it is also able to convert a range of amide substrates into ammonia via its AmiE amidase and AmiF formamidase enzymes. Here data are provided that demonstrate that the iron-responsive regulatory protein Fur directly and indirectly regulates the activity of the two H. pylori amidases. In contrast to other amidase-positive bacteria, amidase and formamidase enzyme activities were not induced by medium supplementation with their respective substrates, acrylamide and formamide. AmiE protein expression and amidase enzyme activity were iron-repressed in H. pylori 26695 but constitutive in the isogenic fur mutant. This regulation was mediated at the transcriptional level via the binding of Fur to the amiE promoter region. In contrast, formamidase enzyme activity was not iron-repressed but was significantly higher in the fur mutant. This effect was not mediated at the transcriptional level, and Fur did not bind to the amiF promoter region. These roles of Fur in regulation of the H. pylori amidases suggest that the H. pylori Fur regulator may have acquired extra functions to compensate for the absence of other regulatory systems.


Asunto(s)
Amidohidrolasas/biosíntesis , Amoníaco/metabolismo , Proteínas Bacterianas/metabolismo , Regulación de la Expresión Génica , Helicobacter pylori/metabolismo , Proteínas Reguladoras del Hierro/metabolismo , Proteínas Represoras/metabolismo , Amidohidrolasas/metabolismo , Proteínas Bacterianas/química , Secuencia de Bases , ADN/metabolismo , Helicobacter pylori/patogenicidad , Humanos , Hierro/farmacología , Modelos Biológicos , Datos de Secuencia Molecular , Hibridación de Ácido Nucleico , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , ARN/metabolismo , Proteínas Represoras/química , Especificidad por Sustrato , Transcripción Genética , Ureasa/química
20.
Helicobacter ; 7(4): 237-44, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12165031

RESUMEN

BACKGROUND: Availability of the essential nutrient iron is thought to vary greatly in the gastric mucosa, and thus the human gastric pathogen Helicobacter pylori requires regulatory responses to these environmental changes. Bacterial iron-responsive regulation is often mediated by Ferric Uptake Regulator (Fur) homologs, and in this study we have determined the role of H. pylori Fur in regulation of H. pylori iron uptake. METHODS: Wild-type H. pylori and fur mutant derivatives were compared after growth in iron-restricted and iron-replete conditions. Iron-uptake was measured using 55Fe-labeled iron, whereas gene expression was monitored at the transcriptional level using Northern hybridization and lacZ reporter gene fusions. RESULTS: Iron-uptake and total cellular iron content were approximately five-fold increased in the fur mutant compared with the wild-type strain, which indicated that in the fur mutant iron-uptake is not repressed by excess iron. A comprehensive screening of all H. pylori genes encoding putative iron-uptake proteins indicated that some of these H. pylori genes are constitutively expressed, while others are iron- and Fur-regulated. CONCLUSIONS: Iron uptake in H. pylori is in part differently regulated compared with other bacteria, since in H. pylori some iron-uptake systems are constitutively expressed. However, other iron uptake systems of H. pylori display the iron- and Fur-mediated repression that is common in bacteria. Taken together, this Fur-mediated modulation of iron-uptake capacity may be a specific adaptation to the conditions in the human stomach, where iron starvation and iron overload can be encountered in relatively short time intervals.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas de Escherichia coli , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/metabolismo , Hierro/metabolismo , Receptores de Superficie Celular , Proteínas Represoras/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Transporte Biológico , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Medios de Cultivo , Helicobacter pylori/genética , Helicobacter pylori/crecimiento & desarrollo , Humanos , Operón Lac , Mutación , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión , Proteínas Represoras/genética , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...