Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Front Microbiol ; 13: 929194, 2022.
Article in English | MEDLINE | ID: mdl-36090072

ABSTRACT

One important factor that promotes the colonization of the upper digestive system of the human pathogen Helicobacter pylori is its helical cell shape. The bacteria cell shape is predominantly defined by its peptidoglycan cell wall. In rod-shaped species, PG synthesis is mediated by two dynamic molecular machines that facilitate growth along the perpendicular axis and the septum, called the elongasome and the divisome, respectively. Furthermore, many bacteria evolved additional mechanisms to locally change PG synthesis patterns to generate diverse cell shapes. Recent work characterizing cell shape mutants of Helicobacter pylori revealed a novel mechanism for the generation of a twisted helix from a rod, including PG-modifying enzymes as well as additional proteins such as the bactofilin homolog CcmA or the membrane proteins Csd5 and Csd7. In this study, we investigate the localization and dynamics of CcmA and Csd7 using live-cell imaging. We also address the question of how these change in the presence or absence of the putative interaction partners.

2.
PLoS One ; 14(6): e0218474, 2019.
Article in English | MEDLINE | ID: mdl-31233532

ABSTRACT

The human pathogen Helicobacter pylori is known for its colonization of the upper digestive system, where it escapes the harsh acidic environment by hiding in the mucus layer. One factor promoting this colonization is the helical cell shape of H. pylori. Among shape determining proteins are cytoskeletal elements like the recently discovered bactofilins. Bactofilins constitute a widespread family of polymer-forming bacterial proteins whose biology is still poorly investigated. Here we describe the first biochemical analysis of the bactofilin HP1542 of H. pylori reference strain 26695. Purified HP1542 forms sheet-like 2D crystalline assemblies, which clearly depend on a natively structured C-terminus. Polymerization properties and protein stability were investigated. Additionally, we also could demarcate HP1542 from amyloid proteins that share similarities with the bactofilin DUF domain. By using zonal centrifugation of total H. pylori cell lysates and immunfluorescence analysis we revealed peripheral membrane association of HP1542 mostly pronounced near mid-cell. Interestingly our results indicate that H. pylori bactofilin does not contribute to cell wall stability. This study might act as a starting point for biophysical studies of the H. pylori bactofilin biology as well as for the investigation of bactofilin cell physiology in this organism. Importantly, this study is the first biochemical analysis of a bactofilin in a human pathogen.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Biochemical Phenomena , Helicobacter pylori/metabolism , Bacterial Proteins/isolation & purification , Bacterial Proteins/ultrastructure , Cell Wall/chemistry , Cell Wall/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Recombinant Proteins , Spectrum Analysis , Structure-Activity Relationship
3.
PLoS One ; 10(3): e0121463, 2015.
Article in English | MEDLINE | ID: mdl-25822999

ABSTRACT

Pathogenicity of the human pathogen Helicobacter pylori relies on its capacity to adapt to a hostile environment and to escape the host response. Although there have been great advances in our understanding of the bacterial cytoskeleton, major gaps remain in our knowledge of its contribution to virulence. In this study we have explored the influence of coiled coil rich proteins (Ccrp) cytoskeletal elements on pathogenicity factors of H. pylori. Deletion of any of the ccrp resulted in a strongly decreased activity of the main pathogenicity factor urease. We further investigated their role using in vitro co-culture experiments with the human gastric adenocarcinoma cell line AGS modeling H. pylori - host cell interactions. Intriguingly, host cell showed only a weak "scattering/hummingbird" phenotype, in which host cells are transformed from a uniform polygonal shape into a severely elongated state characterized by the formation of needle-like projections, after co-incubation with any ccrp deletion mutant. Furthermore, co-incubation with the ccrp59 mutant resulted in reduced type IV secretion system associated activities, e.g. IL-8 production and CagA translocation/phosphorylation. Thus, in addition to their role in maintaining the helical cell shape of H. pylori Ccrp proteins influence many cellular processes and are thereby crucial for the virulence of this human pathogen.


Subject(s)
Bacterial Proteins/physiology , Helicobacter pylori/pathogenicity , Antigens, Bacterial/physiology , Bacterial Proteins/genetics , Cell Line , Genes, Bacterial , Helicobacter Infections/etiology , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Helicobacter pylori/physiology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Interleukin-8/biosynthesis , Mutation , Phenotype , Type IV Secretion Systems/genetics , Type IV Secretion Systems/physiology , Urease/metabolism , Virulence/genetics , Virulence/physiology , Virulence Factors/genetics , Virulence Factors/physiology
4.
J Bacteriol ; 195(7): 1411-20, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23335414

ABSTRACT

Of the various kinds of cell division, the most common mode is binary fission, the division of a cell into two morphologically identical daughter cells. However, in the case of asymmetric cell division, Caulobacter crescentus produces two morphologically and functionally distinct cell types. Here, we have studied cell cycle progression of the human pathogen Helicobacter pylori using a functional green fluorescent protein (GFP) fusion of FtsZ protein and membrane staining. In small cells, representing newly divided cells, FtsZ localizes to a single cell pole. During the cell cycle, spiral intermediates are formed until an FtsZ ring is positioned with very little precision, such that central as well as acentral rings can be observed. Daughter cells showed considerably different sizes, suggesting that H. pylori divides asymmetrically. Fluorescence recovery after photobleaching (FRAP) analyses demonstrate that the H. pylori FtsZ ring is about as dynamic as that of Escherichia coli but that polar assemblies show less turnover. Strikingly, our results demonstrate that H. pylori cell division follows a different route from that in E. coli and Bacillus subtilis. It is also different from that in C. crescentus, where cytokinesis regulation proteins like MipZ play a role. Therefore, this report provides the first cell-biological analysis of FtsZ dynamics in the human pathogen H. pylori and even in epsilonproteobacteria to our knowledge. In addition, analysis of the filament architecture of H. pylori and E. coli FtsZ filaments in the heterologous system of Drosophila melanogaster S2 Schneider cells revealed that both have different filamentation properties in vivo, suggesting a unique intrinsic characteristic of each protein.


Subject(s)
Bacterial Proteins/analysis , Cell Division , Cytoskeletal Proteins/analysis , Helicobacter pylori/chemistry , Helicobacter pylori/physiology , Bacterial Proteins/genetics , Cytoskeletal Proteins/genetics , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Helicobacter pylori/cytology , Helicobacter pylori/metabolism , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics
5.
Int J Syst Evol Microbiol ; 63(Pt 4): 1499-1504, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22863988

ABSTRACT

A Gram-stain-negative, non-motile, facultatively anaerobic, acid-tolerant rod, designated strain DKE6(T), was isolated from an acidic biofilm (pH 2.5) harvested in the pyrite mine Drei Kronen und Ehrt in Germany. The isolate grew optimally at pH 5.5, between 25 and 30 °C and only with casein as the carbon and energy source; although a variety of sugars were tested as growth substrates, none supported growth of the isolate. During casein consumption, strain DKE6(T) produced ammonium, which led to an alkalinization of the medium. This is a possible strategy to raise the pH in the direct vicinity of the cell and hence modulate the pH towards the growth optimum. The predominant fatty acids (>5 %) were iso-C11 : 0 3-OH, iso-C15 : 0, iso-C17 : 0 and iso-C17 : 1ω9c. The DNA G+C content was 66.6 %. Strain DKE6(T) was not able to oxidize iron or thiosulfate. Iron reduction was detected. The isolate showed 93.3 % 16S rRNA gene sequence similarity to the most closely related cultivable strain, Dokdonella koreensis DS-123(T), but <93.2 % sequence similarity with other type strains of closely related type species of the Gammaproteobacteria. On the basis of physiological and biochemical data, the isolate is considered to represent a novel species of a new genus in the class Gammaproteobacteria, for which we propose the name Metallibacterium scheffleri gen. nov., sp. nov. The type strain of the type species is DKE6(T) ( = DSM 24874(T) = JCM 17596(T)).


Subject(s)
Mining , Phylogeny , Xanthomonadaceae/classification , Bacterial Typing Techniques , Base Composition , Biofilms , Cytochromes/analysis , DNA, Bacterial/genetics , Fatty Acids/analysis , Germany , Iron , Molecular Sequence Data , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Sulfides , Ubiquinone/analysis , Xanthomonadaceae/genetics , Xanthomonadaceae/isolation & purification
6.
J Bacteriol ; 193(17): 4523-30, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21642462

ABSTRACT

We identified two additional genes of Helicobacter pylori encoding Ccrp proteins. All four Ccrps have different multimerization and filamentation properties and different types of smallest subunits and do not copurify, suggesting a system of individual Ccrp filaments. Despite the presence of morphologically unaltered flagella, all ccrp mutants displayed significantly reduced motility.


Subject(s)
Bacterial Proteins/metabolism , Genes, Bacterial , Helicobacter pylori/genetics , Bacterial Proteins/genetics , Flagella/metabolism , Gene Expression Regulation, Bacterial , Helicobacter pylori/metabolism , Hydrogen-Ion Concentration , Microscopy, Electron , Plasmids
7.
J Bacteriol ; 192(24): 6378-89, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20952576

ABSTRACT

Cytochrome oxidases are perfect model substrates for analyzing the assembly of multisubunit complexes because the need for cofactor incorporation adds an additional level of complexity to their assembly. cbb(3)-type cytochrome c oxidases (cbb(3)-Cox) consist of the catalytic subunit CcoN, the membrane-bound c-type cytochrome subunits CcoO and CcoP, and the CcoQ subunit, which is required for cbb(3)-Cox stability. Biogenesis of cbb(3)-Cox proceeds via CcoQP and CcoNO subcomplexes, which assemble into the active cbb(3)-Cox. Most bacteria expressing cbb(3)-Cox also contain the ccoGHIS genes, which encode putative cbb(3)-Cox assembly factors. Their exact function, however, has remained unknown. Here we analyzed the role of CcoH in cbb(3)-Cox assembly and showed that CcoH is a single spanning-membrane protein with an N-terminus-out-C-terminus-in (N(out)-C(in)) topology. In its absence, neither the fully assembled cbb(3)-Cox nor the CcoQP or CcoNO subcomplex was detectable. By chemical cross-linking, we demonstrated that CcoH binds primarily via its transmembrane domain to the CcoP subunit of cbb(3)-Cox. A second hydrophobic stretch, which is located at the C terminus of CcoH, appears not to be required for contacting CcoP, but deleting it prevents the formation of the active cbb(3)-Cox. This suggests that the second hydrophobic domain is required for merging the CcoNO and CcoPQ subcomplexes into the active cbb(3)-Cox. Surprisingly, CcoH does not seem to interact only transiently with the cbb(3)-Cox but appears to stay tightly associated with the active, fully assembled complex. Thus, CcoH behaves more like a bona fide subunit of the cbb(3)-Cox than an assembly factor per se.


Subject(s)
Electron Transport Complex IV/metabolism , Escherichia coli/metabolism , Protein Subunits/metabolism , Rhodobacter capsulatus/metabolism , Bacterial Proteins/metabolism , Cell Respiration/physiology , Electron Transport Complex IV/genetics , Gene Expression Regulation, Bacterial/physiology , Plasmids , Protein Conformation , Protein Subunits/chemistry , Rhodobacter capsulatus/genetics
10.
PLoS Pathog ; 5(11): e1000669, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19936218

ABSTRACT

Pathogenicity of the human pathogen Helicobacter pylori relies upon its capacity to adapt to a hostile environment and to escape from the host response. Therefore, cell shape, motility, and pH homeostasis of these bacteria are specifically adapted to the gastric mucus. We have found that the helical shape of H. pylori depends on coiled coil rich proteins (Ccrp), which form extended filamentous structures in vitro and in vivo, and are differentially required for the maintenance of cell morphology. We have developed an in vivo localization system for this pathogen. Consistent with a cytoskeleton-like structure, Ccrp proteins localized in a regular punctuate and static pattern within H. pylori cells. Ccrp genes show a high degree of sequence variation, which could be the reason for the morphological diversity between H. pylori strains. In contrast to other bacteria, the actin-like MreB protein is dispensable for viability in H. pylori, and does not affect cell shape, but cell length and chromosome segregation. In addition, mreB mutant cells displayed significantly reduced urease activity, and thus compromise a major pathogenicity factor of H. pylori. Our findings reveal that Ccrp proteins, but not MreB, affect cell morphology, while both cytoskeletal components affect the development of pathogenicity factors and/or cell cycle progression.


Subject(s)
Bacterial Proteins/metabolism , Cytoskeleton/metabolism , Helicobacter pylori/metabolism , Cell Cycle/physiology , Cell Movement/physiology , Cell Shape/physiology , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Hydrogen-Ion Concentration , Urease/metabolism
11.
J Bacteriol ; 187(13): 4683-8, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15968080

ABSTRACT

Here we describe that the Helicobacter pylori sensor kinase produced by HP1364 and the response regulator produced by HP1365 and designated CrdS and CrdR, respectively, are both required for transcriptional induction of the H. pylori copper resistance determinant CrdA by copper ions. CrdRS-deficient mutants lacked copper induction of crdA expression and were copper sensitive. A direct role of CrdR in transcriptional regulation of crdA was confirmed by in vitro binding of CrdR to the crdA upstream region. A 21-nucleotide sequence located near the crdA promoter was shown to be required for CrdR binding.


Subject(s)
Bacterial Proteins/genetics , Copper/pharmacology , Genes, Bacterial/genetics , Helicobacter pylori/drug effects , Protein Kinases/genetics , Trans-Activators/genetics , Base Sequence , Binding Sites/genetics , Drug Resistance, Bacterial/genetics , Helicobacter pylori/genetics , Molecular Sequence Data , Promoter Regions, Genetic , Protein Kinases/metabolism , Transcription, Genetic
12.
J Bacteriol ; 187(11): 3687-92, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15901691

ABSTRACT

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.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Helicobacter pylori/genetics , Iron/metabolism , Oxidative Stress/physiology , Repressor Proteins/metabolism , Superoxide Dismutase/genetics , Base Sequence , DNA Footprinting , Gene Expression Regulation, Bacterial , Helicobacter pylori/metabolism , Promoter Regions, Genetic/physiology , Superoxide Dismutase/metabolism , Superoxides/metabolism , Transcription, Genetic
13.
Microbiology (Reading) ; 151(Pt 2): 533-546, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15699202

ABSTRACT

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.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Helicobacter pylori/metabolism , Iron/metabolism , Repressor Proteins/metabolism , Bacterial Proteins/genetics , Base Sequence , Helicobacter pylori/genetics , Helicobacter pylori/growth & development , Humans , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Proteome , Repressor Proteins/genetics , Transcription, Genetic
14.
Helicobacter ; 9 Suppl 1: 15-22, 2004.
Article in English | MEDLINE | ID: mdl-15347301

ABSTRACT

Research in the last year has provided new insights into the function of the the cag-associated type IV secretion system and the vacuolating toxin VacA. A quite new aspect was disclosed by the finding that Helicobacter pylori in Mongolian gerbils colonizes a very distinct topology in the gastric mucous layer, obviously providing optimal conditions for long-term survival. Further research activities focused on H. pylori ammonia and metal metabolism as well as on bacterial stress defence mechanisms. Differential expression of approximately 7% of the bacterial genome was found at low pH suggesting that H. pylori has evolved a multitude of acid-adaptive mechanisms. VacA was shown to interrupt phagosome maturation in macrophage cell lines as well as to modulate and interfere with T lymphocyte immunological functions. Gastric mucosa as well as the H. pylori-infected epithelial cell line AGS strongly express IL-8 receptor A and B, which might contribute to the augmentation of the inflammatory response. Accumulating evidence implicates genetic variation in the inflammatory response to H. pylori in the etiology of the increased risk of gastric cancer after H. pylori infection. The chronic imbalance between apoptosis and cell proliferation is the first step of gastric carcinogenesis. In this regard, it was demonstrated that coexpression of two H. pylori proteins, CagA and HspB, in AGS cells, caused an increase in E2F transcription factor, cyclin D3, and phosphorylated retinoblastoma protein. Taken together, we now have a better understanding of the role of different virulence factors of H. pylori. There is still a lot to be learned, but the promising discoveries summarized here, demonstrate that the investigation of the bacterial survival strategies will give novel insights into pathogenesis and disease development.


Subject(s)
Helicobacter Infections/complications , Helicobacter Infections/microbiology , Helicobacter pylori/physiology , Virulence Factors/physiology , Adaptation, Physiological , Animals , Bacterial Proteins/metabolism , Disease Models, Animal , Gastric Mucosa/microbiology , Gene Expression Regulation, Bacterial/physiology , Heat-Shock Proteins/metabolism , Helicobacter pylori/pathogenicity , Metals/metabolism , Stomach Neoplasms/etiology , Virulence Factors/metabolism
15.
J Biol Chem ; 278(9): 6905-11, 2003 Feb 28.
Article in English | MEDLINE | ID: mdl-12486029

ABSTRACT

Poxviruses have evolved various strategies to counteract the host immune response, one of which is based on the expression of soluble cytokine receptors. Using various biological assays, we detected a chicken interferon-gamma (chIFN-gamma)-neutralizing activity in supernatants of fowlpox virus (FPV)-infected cells that could be destroyed by trypsin treatment. Secreted viral proteins were purified by affinity chromatography using matrix-immobilized chIFN-gamma, followed by two-dimensional gel electrophoresis. Matrix-assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF MS) analysis indicated that the viral IFN-gamma-binding protein in question was encoded by the FPV gene 016. The chicken IFN-gamma binding and neutralizing activity of the recombinant FPV016 protein was confirmed using supernatants of cells infected with a recombinant vaccinia virus that lacked its own IFN-gamma-binding protein but instead expressed the FPV016 gene. The FPV016 gene product also neutralized the activity of duck and human IFN-gamma but failed to neutralize the activity of mouse and rat IFN-gamma. Unlike previously known cellular and poxviral IFN-gamma receptors, which all contain fibronectin type III domains, the IFN-gamma-binding protein of FPV contains an immunoglobulin domain. Remarkably, it exhibits no significant homology to any known viral or cellular protein. Because IFN-gamma receptors of birds have not yet been characterized at the molecular level, the possibility remains that FPV016 represents a hijacked chicken gene and that avian and mammalian IFN-gamma receptors have fundamentally different primary structures.


Subject(s)
Carrier Proteins/genetics , Fowlpox virus/genetics , Interferon-gamma/metabolism , Viral Proteins/genetics , Amino Acid Sequence , Animals , COS Cells , Carrier Proteins/chemistry , Chickens , Chromatography , Electrophoresis, Gel, Two-Dimensional , Fibroblasts/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Isoelectric Focusing , Mice , Molecular Sequence Data , Nitric Oxide/metabolism , Protein Binding , Protein Structure, Tertiary , Quail , RNA/metabolism , Rats , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Time Factors , Trypsin/pharmacology , Vaccinia virus/genetics , Viral Proteins/chemistry
16.
J Bacteriol ; 184(23): 6700-8, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12426358

ABSTRACT

Mechanisms involved in maintaining cytoplasmic metal ion homeostasis play a central role in the adaptation of Helicobacter pylori to the changing gastric environment. An investigation of the global regulatory responses to copper ions by using RNA profiling with a threshold factor of 4.0 revealed that copper induces transcription of 19 H. pylori genes and that only the ferritin gene pfr is repressed. The 57-fold copper induction identified the HP1326 gene encoding an H. pylori-specific protein as a candidate for a novel copper resistance determinant. The HP1326 gene is expressed as a monocistronic unit, and two small HP1326 mRNAs are copper induced. The HP1326 protein is secreted and is required for copper resistance maintained by cytoplasmic copper homeostasis, as H. pylori HP1326 mutants were copper sensitive and displayed increased copper induction of HP1326 transcription as well as elevated copper repression of ferritin synthesis. The clear copper-sensitive phenotype displayed by H. pylori HP1327 and HP1328 mutants provides strong evidence that the HP1326 protein, together with the signal peptide site of the H. pylori-specific protein HP1327, whose gene is located downstream from that encoding HP1326, and the CzcB and CzcA metal efflux system component homologs HP1328 and HP1329, constitutes a novel type of copper efflux pump, as discussed below. The HP1329 gene could not be inactivated, but the 14-fold transcriptional copper induction determined by RNA profiling points towards a function of the encoded CzcA homolog in copper resistance. In summary, results from RNA profiling identified the novel H. pylori-specific copper resistance determinants CrdA (HP1326) and CrdB (HP1327), which are required for adaptation to copper-rich environmental conditions.


Subject(s)
Cation Transport Proteins/genetics , Copper/pharmacology , DNA Mutational Analysis , Gene Expression Profiling , Helicobacter pylori/drug effects , Oligonucleotide Array Sequence Analysis , RNA, Bacterial/genetics , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Cation Transport Proteins/metabolism , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial , Helicobacter pylori/genetics , Helicobacter pylori/growth & development , Molecular Sequence Data , RNA, Bacterial/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
Infect Immun ; 70(7): 3930-4, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12065537

ABSTRACT

We show here that Mg(2+) acquisition by CorA is essential for Helicobacter pylori in vitro, as corA mutants did not grow in media without Mg(2+) supplementation. Complementation analysis performed with an Escherichia coli corA mutant revealed that H. pylori CorA transports nickel and cobalt in addition to Mg(2+). However, Mg(2+) is the dominant CorA substrate, as the corA mutation affected neither cobalt and nickel resistance nor nickel induction of urease in H. pylori. The drastic Mg(2+) requirement (20 mM) of H. pylori corA mutants indicates that CorA plays a key role in the adaptation to the low-Mg(2+) conditions predominant in the gastric environment.


Subject(s)
Bacterial Proteins/metabolism , Cation Transport Proteins/metabolism , Helicobacter pylori/metabolism , Magnesium/metabolism , Bacterial Proteins/genetics , Cation Transport Proteins/genetics , Cobalt/metabolism , Escherichia coli/metabolism , Helicobacter pylori/genetics , Helicobacter pylori/growth & development , Helicobacter pylori/isolation & purification , Nickel/metabolism , Stomach/microbiology
18.
Infect Immun ; 70(7): 3923-9, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12065536

ABSTRACT

The reactivity of the essential element iron necessitates a concerted expression of ferritins, which mediate iron storage in a nonreactive state. Here we have further established the role of the Helicobacter pylori ferritin Pfr in iron metabolism and gastric colonization. Iron stored in Pfr enabled H. pylori to multiply under severe iron starvation and protected the bacteria from acid-amplified iron toxicity, as inactivation of the pfr gene restricted growth of H. pylori under these conditions. The lowered total iron content in the pfr mutant, which is probably caused by decreased iron uptake rates, was also reflected by an increased resistance to superoxide stress. Iron induction of Pfr synthesis was clearly diminished in an H. pylori feoB mutant, which lacked high-affinity ferrous iron transport, confirming that Pfr expression is mediated by changes in the cytoplasmic iron pool and not by extracellular iron. This is well in agreement with the recent discovery that iron induces Pfr synthesis by abolishing Fur-mediated repression of pfr transcription, which was further confirmed here by the observation that iron inhibited the in vitro binding of recombinant H. pylori Fur to the pfr promoter region. The functions of H. pylori Pfr in iron metabolism are essential for survival in the gastric mucosa, as the pfr mutant was unable to colonize in a Mongolian gerbil-based animal model. In summary, the pfr phenotypes observed give new insights into prokaryotic ferritin functions and indicate that iron storage and homeostasis are of extraordinary importance for H. pylori to survive in its hostile natural environment.


Subject(s)
Bacterial Proteins/physiology , Ferritins/analogs & derivatives , Ferritins/physiology , Helicobacter pylori/metabolism , Iron/metabolism , Stomach/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Disease Models, Animal , Ferritins/genetics , Gerbillinae , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Helicobacter pylori/growth & development , Hydrogen-Ion Concentration , Mutagenesis , Oxidative Stress , Paraquat/pharmacology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Superoxides/metabolism
19.
Infect Immun ; 70(2): 606-11, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11796589

ABSTRACT

The only known niche of the human pathogen Helicobacter pylori is the gastric mucosa, where large fluctuations of pH occur, indicating that the bacterial response and resistance to acid are important for successful colonization. One of the few regulatory proteins in the H. pylori genome is a homologue of the ferric uptake regulator (Fur). In most bacteria, the main function of Fur is the regulation of iron homeostasis. However, in Salmonella enterica serovar Typhimurium, Fur also plays an important role in acid resistance. In this study, we determined the role of the H. pylori Fur homologue in acid resistance. Isogenic fur mutants were generated in three H. pylori strains (1061, 26695, and NCTC 11638). At pH 7 there was no difference between the growth rates of mutants and the parent strains. Under acidic conditions, growth of the fur mutants was severely impaired. No differences were observed between the survival of the fur mutant and parent strain 1061 after acid shock. Addition of extra iron or removal of iron from the growth medium did not improve the growth of the fur mutant at acidic pH. This indicates that the phenotype of the fur mutant at low pH was not due to increased iron sensitivity. Transcription of fur was repressed in response to low pH. From this we conclude that Fur is involved in the growth at acidic pH of H. pylori; as such, it is the first regulatory protein implicated in the acid resistance of this important human pathogen.


Subject(s)
Bacterial Proteins/physiology , Helicobacter pylori/drug effects , Hydrochloric Acid/pharmacology , Repressor Proteins/physiology , Acids/pharmacology , Bacterial Proteins/genetics , Culture Media , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial/drug effects , Helicobacter pylori/genetics , Helicobacter pylori/growth & development , Hydrogen-Ion Concentration , Iron/pharmacology , Mutagenesis , Repressor Proteins/genetics , Transcription, Genetic/drug effects , Urease/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...