Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
PLoS Pathog ; 15(1): e1007538, 2019 01.
Article in English | MEDLINE | ID: mdl-30608981

ABSTRACT

Staphylococcus aureus causes acute and chronic infections resulting in significant morbidity. Urease, an enzyme that generates NH3 and CO2 from urea, is key to pH homeostasis in bacterial pathogens under acidic stress and nitrogen limitation. However, the function of urease in S. aureus niche colonization and nitrogen metabolism has not been extensively studied. We discovered that urease is essential for pH homeostasis and viability in urea-rich environments under weak acid stress. The regulation of urease transcription by CcpA, Agr, and CodY was identified in this study, implying a complex network that controls urease expression in response to changes in metabolic flux. In addition, it was determined that the endogenous urea derived from arginine is not a significant contributor to the intracellular nitrogen pool in non-acidic conditions. Furthermore, we found that during a murine chronic renal infection, urease facilitates S. aureus persistence by promoting bacterial fitness in the low-pH, urea-rich kidney. Overall, our study establishes that urease in S. aureus is not only a primary component of the acid response network but also an important factor required for persistent murine renal infections.


Subject(s)
Staphylococcus aureus/metabolism , Urease/metabolism , Urease/physiology , Acids/metabolism , Animals , Bacterial Proteins/metabolism , Female , Homeostasis/physiology , Hydrogen-Ion Concentration , Kidney/microbiology , Kidney Diseases/microbiology , Male , Mice , Mice, Inbred C57BL , Nitrogen/metabolism , Staphylococcal Infections/metabolism , Staphylococcus aureus/pathogenicity , Urea/metabolism , Urease/genetics
2.
Mol Biol Rep ; 46(1): 133-141, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30374769

ABSTRACT

Urease is a potent metalloenzyme with diverse applications. This paper describes the scale up and purification of an extracellular urease from Arthrobacter creatinolyticus MTCC 5604. The urease production was scaled-up in 3.7 L and 20 L fermentor. A maximum activity of 27 and 27.8 U/mL and a productivity of 0.90 and 0.99 U/mL/h were obtained at 30 h and 28 h in 3.7 and 20 L fermentor, respectively. Urease was purified to homogeneity with 49.85-fold purification by gel filtration and anion exchange chromatography with a yield of 36% and a specific activity of 1044.37 U/mg protein. The enzyme showed three protein bands with molecular mass of 72.6, 11.2 and 6.1 kDa on SDS-PAGE and ~ 270 kDa on native PAGE. The cytotoxic effect of urease was assessed in vitro using cancer cell lines (A549 and MG-63) and normal cell line (HEK 293). Urease showed its inhibitory effects on cancer cell lines through the generation of toxic ammonia, which in turn increased the pH of the surrounding medium. This increase in extracellular pH, enhanced the cytotoxic effect of weak base chemotherapeutic drugs, doxorubicin (50 µM) and vinblastine (100 µM) in the presence of urease (5 U/mL) and urea (0-4 mM) significantly.


Subject(s)
Arthrobacter/enzymology , Urease/isolation & purification , Urease/pharmacology , A549 Cells/drug effects , Ammonia/metabolism , Arthrobacter/metabolism , Arthrobacter/physiology , Cell Line , Chromatography, Gel/methods , Electrophoresis, Polyacrylamide Gel/methods , HEK293 Cells , Humans , Hydrogen-Ion Concentration/drug effects , Molecular Weight , Urea/metabolism , Urease/physiology
3.
Mol Biol Rep ; 46(1): 51-58, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30350237

ABSTRACT

Urease is an important virulence factor for a variety of pathogenic bacteria strains such as Helicobacter pylori, which colonizes human gastric mucosa, and Proteus sp., responsible for urinary tract infections. Specific inhibition of urease activity could be a promising adjuvant strategy for eradication of these pathogens. Due to the interesting antiureolytic activity of carvone and the scant information regarding the inhibitory properties of corresponding monoterpenes, we decided to study selected monoterpenic ketones and their oxygen derivatives. Several monoterpenes and their terpenoid oxygen derivatives were evaluated in vitro against Sporosarcina pasteurii urease. The most effective inhibitors-derivatives of ß-cyclocitral (ester 10 and bromolactone 14)-were described with [Formula: see text] of 46.7 µM and 45.8 µM, respectively. Active inhibitors of native urease were tested against H. pylori and Proteus mirabilis whole cells. Here, the most active inhibitor, 14, was characterized with IC50 values of 0.32 mM and 0.61 mM for P. mirabilis and H. pylori, respectively. The antibacterial activity of a few tested inhibitors was also observed. Compound 14 limited the growth of E. coli ([Formula: see text]= 250 µg/mL). Interestingly, 10 was the only compound that was effective against both Gram-negative and Gram-positive bacteria. It had a [Formula: see text] of 150 µg/mL against E. coli and S. aureus. In the presented study a group of novel antiureolytic compounds was characterised. Besides carvone stereoisomers, these are the only terpenoid urease inhibitors described so far.


Subject(s)
Terpenes/pharmacology , Urease/antagonists & inhibitors , Urinary Tract Infections/drug therapy , Aldehydes/pharmacology , Anti-Bacterial Agents/pharmacology , Diterpenes/pharmacology , Escherichia coli/drug effects , Gastric Mucosa/drug effects , Helicobacter pylori/drug effects , Helicobacter pylori/pathogenicity , Humans , Monoterpenes , Plant Extracts/pharmacology , Sporosarcina/drug effects , Sporosarcina/pathogenicity , Staphylococcus aureus/drug effects , Urease/physiology
4.
PLoS Biol ; 16(1): e2003887, 2018 01.
Article in English | MEDLINE | ID: mdl-29320492

ABSTRACT

Urease as a potential target of antimicrobial drugs has received considerable attention given its versatile roles in microbial infection. Development of effective urease inhibitors, however, is a significant challenge due to the deeply buried active site and highly specific substrate of a bacterial urease. Conventionally, urease inhibitors are designed by either targeting the active site or mimicking substrate of urease, which is not efficient. Up to now, only one effective inhibitor-acetohydroxamic acid (AHA)-is clinically available, but it has adverse side effects. Herein, we demonstrate that a clinically used drug, colloidal bismuth subcitrate, utilizes an unusual way to inhibit urease activity, i.e., disruption of urease maturation process via functional perturbation of a metallochaperone, UreG. Similar phenomena were also observed in various pathogenic bacteria, suggesting that UreG may serve as a general target for design of new types of urease inhibitors. Using Helicobacter pylori UreG as a showcase, by virtual screening combined with experimental validation, we show that two compounds targeting UreG also efficiently inhibited urease activity with inhibitory concentration (IC)50 values of micromolar level, resulting in attenuated virulence of the pathogen. We further demonstrate the efficacy of the compounds in a mammalian cell infection model. This study opens up a new opportunity for the design of more effective urease inhibitors and clearly indicates that metallochaperones involved in the maturation of important microbial metalloenzymes serve as new targets for devising a new type of antimicrobial drugs.


Subject(s)
Bacterial Proteins/drug effects , Carrier Proteins/drug effects , Organometallic Compounds/pharmacology , Urease/antagonists & inhibitors , Anti-Infective Agents/pharmacology , Bacterial Proteins/physiology , Carrier Proteins/physiology , Catalytic Domain , Helicobacter pylori/metabolism , Metallochaperones/pharmacology , Phosphate-Binding Proteins , Urease/physiology , Virulence
5.
Chem Soc Rev ; 44(21): 7465-83, 2015 Nov 07.
Article in English | MEDLINE | ID: mdl-26214155

ABSTRACT

Living cells are able to produce a wide variety of biological responses when subjected to biochemical stimuli. It has become apparent that these biological responses are regulated by complex chemical reaction networks (CRNs). Unravelling the function of these circuits is a key topic of both systems biology and synthetic biology. Recent progress at the interface of chemistry and biology together with the realisation that current experimental tools are insufficient to quantitatively understand the molecular logic of pathways inside living cells has triggered renewed interest in the bottom-up development of CRNs. This builds upon earlier work of physical chemists who extensively studied inorganic CRNs and showed how a system of chemical reactions can give rise to complex spatiotemporal responses such as oscillations and pattern formation. Using purified biochemical components, in vitro synthetic biologists have started to engineer simplified model systems with the goal of mimicking biological responses of intracellular circuits. Emulation and reconstruction of system-level properties of intracellular networks using simplified circuits are able to reveal key design principles and molecular programs that underlie the biological function of interest. In this Tutorial Review, we present an accessible overview of this emerging field starting with key studies on inorganic CRNs followed by a discussion of recent work involving purified biochemical components. Finally, we review recent work showing the versatility of programmable biochemical reaction networks (BRNs) in analytical and diagnostic applications.


Subject(s)
Models, Biological , Bioengineering , Signal Transduction , Transcriptional Activation , Urea/chemistry , Urease/physiology
8.
Urologe A ; 49(5): 623-8, 2010 May.
Article in German | MEDLINE | ID: mdl-20449779

ABSTRACT

The classic "infection stone" struvite is formed as a result of metabolic activity of urease-positive bacteria from alkaline urine with pH-values above 7.5. Due to improved infection diagnostics and antibiotic therapy, the occurrence of infection-related urinary stones in the western industrialized world decreases, despite the generally increasing prevalence rates of urolithiasis in these societies. Struvite is often associated with other mineral phases. These accessory mineral phases could indicate other, non-infection-related causes of urinary stone formation. Thus, mineral analysis is always recommended. Struvite stones as well as struvite encrustations on urinary tract implants are characterized by rapid growth. The rapid growth-related embedding of urease-positive bacteria in the crystalline material makes the urinary stone a persistent source of recurrent urinary tract infections. According to the German Society of Urology guidelines on urolithiasis, a patient with the diagnosis "infection stone" should be assigned to the "high-risk" patient group. Complete stone and debris removal, as well as a special metaphylaxis strategy are required to initiate successful stone therapy.


Subject(s)
Bacterial Infections/complications , Urinary Calculi/etiology , Urinary Tract Infections/complications , Acid-Base Equilibrium/physiology , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Drug Resistance, Microbial , Humans , Magnesium Compounds/urine , Phosphates/urine , Recurrence , Risk Factors , Struvite , Urease/physiology , Urinary Calculi/chemistry , Urinary Calculi/microbiology , Urinary Calculi/prevention & control , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology
9.
J Clin Pathol ; 63(5): 424-30, 2010 May.
Article in English | MEDLINE | ID: mdl-20418234

ABSTRACT

Urease is known to be a major contributor to pathologies induced by Helicobacter pylori and Proteus species. In H pylori, urease allows the bacteria to survive in an acidic gastric environment during colonisation, playing an important role in the pathogenesis of gastric and peptic ulcers. Ureolytic activity also results in the production of ammonia in close proximity to the gastric epithelium, causing cell damage and inflammation. In the case of Proteus species (notably Proteus mirabilis) infection, stones are formed due to the presence of ammonia and carbon dioxide released by urease action. In addition, the ammonia released is able to damage the glycosaminoglycan layer, which protects the urothelial surface against bacterial infection. In this context, the administration of urease inhibitors may be an effective therapy for urease-dependent pathogenic bacteria. This is a review of the role of ureases in H pylori and Proteus species infections, focussing on the biochemical and clinical aspects of the most promising and/or potent urease inhibitors for the treatment of gastric and urinary tract infections.


Subject(s)
Gastritis/drug therapy , Helicobacter Infections/drug therapy , Proteus Infections/drug therapy , Urease/antagonists & inhibitors , Urinary Tract Infections/drug therapy , Anti-Bacterial Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Gastritis/microbiology , Helicobacter pylori/enzymology , Helicobacter pylori/pathogenicity , Humans , Proteus/enzymology , Proteus/pathogenicity , Urease/physiology , Virulence
10.
Biosci Biotechnol Biochem ; 74(3): 583-9, 2010.
Article in English | MEDLINE | ID: mdl-20208338

ABSTRACT

Urolith, which consists of dirty yellow-colored attachments on the toilet bowl, is associated with a variety of odorous chemicals, including ammonia, and causes disadvantages in daily life. Although largely it is derived from microorganisms, little is known about the microbial processes underlying the formation of urolith. In order to gain insight into the types and the activities of microorganisms present in urolith, culturable bacteria were isolated, identified, and physiologically characterized. One of the isolates exhibited higher ability to produce ammonia when it was grown in artificial urine medium. Phylogenetic and physiological analyses indicated that this strain (T-02) belonged to a new group of Staphylococcus species, showing combined phenotypes as between S. lentus and S. xylosus. T-02 exhibited high urease activity and was capable of growing in the urinary condition by forming robust biofilms. The results of this study indicate that T-02 has successfully adapted itself to the environment of urolith.


Subject(s)
Biofilms , Staphylococcus/physiology , Toilet Facilities , Urease/physiology , Ammonia/metabolism , Phylogeny , Staphylococcus/enzymology , Staphylococcus/isolation & purification , Urine/microbiology
11.
J Cell Mol Med ; 14(7): 2025-34, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19754669

ABSTRACT

The bacterium Helicobacter pylori causes peptic ulcers and gastric cancer in human beings by mechanisms yet not fully understood. H. pylori produces urease which neutralizes the acidic medium permitting its survival in the stomach. We have previously shown that ureases from jackbean, soybean or Bacillus pasteurii induce blood platelet aggregation independently of their enzyme activity by a pathway requiring platelet secretion, activation of calcium channels and lipoxygenase-derived eicosanoids. We investigated whether H. pylori urease displays platelet-activating properties and defined biochemical pathways involved in this phenomenon. For that the effects of purified recombinant H. pylori urease (HPU) added to rabbit platelets were assessed turbidimetrically. ATP secretion and production of lipoxygenase metabolites by activated platelets were measured. Fluorescein-labelled HPU bound to platelets but not to erythrocytes. HPU induced aggregation of rabbit platelets (ED(50) 0.28 microM) accompanied by ATP secretion. No correlation was found between platelet activation and ureolytic activity of HPU. Platelet aggregation was blocked by esculetin (12-lipoxygenase inhibitor) and enhanced approximately 3-fold by indomethacin (cyclooxygenase inhibitor). A metabolite of 12-lipoxygenase was produced by platelets exposed to HPU. Platelet responses to HPU did not involve platelet-activating factor, but required activation of verapamil-inhibitable calcium channels. Our data show that purified H. pylori urease activates blood platelets at submicromolar concentrations. This property seems to be common to ureases regardless of their source (plant or bacteria) or quaternary structure (single, di- or tri-chain proteins). These properties of HPU could play an important role in pathogenesis of gastrointestinal and associated cardiovascular diseases caused by H. pylori.


Subject(s)
Helicobacter pylori/enzymology , Lipoxygenases/metabolism , Platelet Activation/physiology , Urease/physiology , Humans
12.
J Biosci ; 34(4): 503-11, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19920336

ABSTRACT

The urease of the human pathogen, Helicobacter pylori, is essential for pathogenesis. The ammonia produced by the enzyme neutralizes stomach acid; thereby modifying its environment. The dodecameric enzyme complex has high affinity for its substrate, urea. We compared urease sequences and derivative 3D homology model structures from all published Helicobacter genomes and an equal number of genomes belonging to strains of another enteric bacterium, Escherichia coli. We found that the enzyme's architecture adapts to fit its niche. This finding, coupled to a survey of other physiological features responsible for the bacterium's acid resistance, suggests how it copes with pH changes caused by disease onset and progression.


Subject(s)
Bacterial Proteins/chemistry , Helicobacter pylori/enzymology , Urease/chemistry , Amino Acid Sequence , Bacterial Proteins/physiology , Crystallography, X-Ray , Escherichia coli/enzymology , Escherichia coli/genetics , Genome, Bacterial , Helicobacter pylori/genetics , Helicobacter pylori/pathogenicity , Homeostasis , Hydrogen-Ion Concentration , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Alignment , Substrate Specificity , Urea/metabolism , Urease/physiology
13.
Appl Environ Microbiol ; 75(21): 6712-20, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19749068

ABSTRACT

Genomic analysis indicated that Edwardsiella ictaluri encodes a putative urease pathogenicity island containing the products of nine open reading frames, including urea and ammonium transporters. In vitro studies with wild-type E. ictaluri and a ureG::kan urease mutant strain indicated that E. ictaluri is significantly tolerant of acid conditions (pH 3.0) but that urease activity is not required for acid tolerance. Growth studies demonstrated that E. ictaluri is unable to grow at pH 5 in the absence of urea but is able to elevate the environmental pH from pH 5 to pH 7 and grow when exogenous urea is available. Substantial production of ammonia was observed for wild-type E. ictaluri in vitro in the presence of urea at low pH, and optimal activity occurred at pH 2 to 3. No ammonia production was detected for the urease mutant. Proteomic analysis with two-dimensional gel electrophoresis indicated that urease proteins are expressed at both pH 5 and pH 7, although urease activity is detectable only at pH 5. Urease was not required for initial invasion of catfish but was required for subsequent proliferation and virulence. Urease was not required for initial uptake or survival in head kidney-derived macrophages but was required for intracellular replication. Intracellular replication of wild-type E. ictaluri was significantly enhanced when urea was present, indicating that urease plays an important role in intracellular survival and replication, possibly through neutralization of the acidic environment of the phagosome.


Subject(s)
Bacterial Proteins/physiology , Edwardsiella ictaluri/enzymology , Edwardsiella ictaluri/pathogenicity , Ictaluridae/microbiology , Macrophages/microbiology , Urease/physiology , Virulence Factors/physiology , Ammonia/metabolism , Animals , Bacterial Proteins/genetics , Colony Count, Microbial , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Edwardsiella ictaluri/genetics , Enterobacteriaceae Infections/microbiology , Gene Deletion , Hydrogen-Ion Concentration , Kidney/microbiology , Molecular Sequence Data , Sequence Analysis, DNA , Urease/genetics , Virulence Factors/genetics
14.
Infect Immun ; 77(10): 4197-208, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19651862

ABSTRACT

Human brucellosis is caused mainly by Brucella melitensis, which is often acquired by ingesting contaminated goat or sheep milk and cheese. Bacterial factors required for food-borne infection of humans by B. melitensis are poorly understood. In this study, a mouse model of oral infection was characterized to assess the roles of urease, the VirB type IV secretion system, and lipopolysaccharide for establishing infection through the digestive tract. B. melitensis strain 16M was consistently recovered from the mesenteric lymph node (MLN), spleen, and liver beginning at 3 or 7 day postinfection (dpi). In the gut, persistence of the inoculum was observed up to 21 dpi. No inflammatory lesions were observed in the ileum or colon during infection. Mutant strains lacking the ureABC genes of the ure1 operon, virB2, or pmm encoding phosphomannomutase were constructed and compared to the wild-type strain for infectivity through the digestive tract. Mutants lacking the virB2 and pmm genes were attenuated in the spleen (P < 0.05) and MLN (P < 0.001), respectively. The wild-type and mutant strains had similar levels of resistance to low pH and 5 or 10% bile, suggesting that the reduced colonization of mutants was not the result of reduced resistance to acid pH or bile salts. In an in vitro lymphoepithelial cell (M-cell) model, B. melitensis transited rapidly through polarized enterocyte monolayers containing M-like cells; however, transit through monolayers containing only enterocytes was reduced or absent. These results indicate that B. melitensis is able to spread systemically from the digestive tract after infection, most likely through M cells of the mucosa-associated lymphoid tissue.


Subject(s)
Bacterial Proteins/physiology , Brucella melitensis/pathogenicity , Gastrointestinal Tract/microbiology , Lipopolysaccharides/physiology , Membrane Transport Proteins/physiology , O Antigens/physiology , Urease/physiology , Virulence Factors/physiology , Animals , Bacterial Proteins/genetics , Cell Line , Colon/pathology , Colony Count, Microbial , Enterocytes/microbiology , Female , Gene Deletion , Ileum/pathology , Lipopolysaccharides/genetics , Liver/microbiology , Lymph Nodes/microbiology , Membrane Transport Proteins/genetics , Mice , Mice, Inbred BALB C , O Antigens/genetics , Spleen/microbiology , Urease/genetics , Virulence Factors/genetics
15.
Appl Environ Microbiol ; 75(18): 5779-86, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19633120

ABSTRACT

O island 48 (OI-48) of Escherichia coli consists of three functional gene clusters that encode urease, tellurite resistance (Te(r)), and putative adhesins Iha and AIDA-1. The functions of these clusters in enterohemorrhagic E. coli (EHEC) O157:H7 infection are unknown. Deletion mutants for these three regions were constructed and evaluated for their ability to adhere to epithelial cells in vitro and in ligated pig ileal loops. Deletion of the Te(r) gene cluster reduced the ability of the organism to adhere to and form large clusters on IPEC-J2 and HEp-2 cells. Complementation of the mutation by introducing the wild-type ter genes restored adherence and large-cluster formation. Tests in ligated pig ileal loops showed a decrease in colonization by the Te(r)-negative mutant, but the difference was not significant compared to colonization by the wild type (26.4% +/- 21.2% versus 40.1% +/- 19.1%; P = 0.168). The OI-48 aidA gene deletion had no effect on adherence in vitro or in vivo. Deletion of the iha and ureC genes had no effect on adherence in vitro but significantly reduced the colonization of EHEC O157:H7 in the ligated pig intestine. These data suggest that Te(r), Iha, and urease may contribute to EHEC O157:H7 pathogenesis by promoting adherence of the pathogen to the host intestinal epithelium.


Subject(s)
Bacterial Adhesion , Epithelial Cells/microbiology , Escherichia coli O157/pathogenicity , Genomic Islands , Ileum/microbiology , Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/physiology , Animals , Cell Line , Escherichia coli O157/genetics , Gene Deletion , Genes, Bacterial , Genetic Complementation Test , Humans , In Vitro Techniques , Multigene Family , Swine , Urease/genetics , Urease/physiology
16.
J Bacteriol ; 191(15): 4798-806, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19502406

ABSTRACT

The citrulline ureidase (CTU) activity has been shown to be associated with highly virulent Francisella tularensis strains, including Schu S4, while it is absent in avirulent or less virulent strains. A definitive role of the ctu gene in virulence and pathogenesis of F. tularensis Schu S4 has not been assessed; thus, an understanding of the significance of this phenotype is long overdue. CTU is a carbon-nitrogen hydrolase encoded by the citrulline ureidase (ctu) gene (FTT0435) on the F. tularensis Schu S4 genome. In the present study, we evaluated the contribution of the ctu gene in the virulence of category A agent F. tularensis Schu S4 by generating a nonpolar deletion mutant, the Deltactu mutant. The deletion of the ctu gene resulted in loss of CTU activity, which was restored by transcomplementing the ctu gene. The Deltactu mutant did not exhibit any growth defect under acellular growth conditions; however, it was impaired for intramacrophage growth in resting as well as gamma interferon-stimulated macrophages. The Deltactu mutant was further tested for its virulence attributes in a mouse model of respiratory tularemia. Mice infected intranasally with the Deltactu mutant showed significantly reduced bacterial burden in the lungs, liver, and spleen compared to wild-type (WT) Schu S4-infected mice. The reduced bacterial burden in mice infected with the Deltactu mutant was also associated with significantly lower histopathological scores in the lungs. Mice infected with the Deltactu mutant succumbed to infection, but they survived longer and showed significantly extended median time to death compared to that shown by WT Schu S4-infected mice. To conclude, this study demonstrates that ctu contributes to intracellular survival, in vivo growth, and pathogenesis. However, ctu is not an absolute requirement for the virulence of F. tularensis Schu S4 in mice.


Subject(s)
Bacterial Proteins/physiology , Citrulline/metabolism , Francisella tularensis/enzymology , Francisella tularensis/pathogenicity , Urease/physiology , Virulence/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Cells, Cultured , Female , Francisella tularensis/genetics , Francisella tularensis/growth & development , Genetic Complementation Test , Lung/microbiology , Macrophages/microbiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Sequence Deletion , Urease/genetics , Urease/metabolism
17.
Gastroenterology ; 136(1): 236-46, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18996125

ABSTRACT

BACKGROUND & AIMS: Helicobacter pylori-induced gastritis predisposes to the development of gastric cancer. Increased epithelial tight junction permeability and alterations in apical-junctional complexes are also associated with an increased risk of carcinogenesis. Phosphorylation of myosin regulatory light chain (MLC) by MLC kinase (MLCK) regulates tight junction function. We determined whether MLCK was activated by H pylori and defined the mechanisms through which such activation dysregulates gastric epithelial barrier function. METHODS: MKN28 gastric epithelial cells were cocultured with the H pylori cag(+) strain 60190 or cagA(-), cagE(-), ureB(-), or vacA(-) mutants. MLC phosphorylation and barrier integrity were determined by immunoblot analysis and transepithelial electrical resistance measurements, respectively. Localization of the tight junction protein occludin was determined by immunocytochemistry in MKN28 cells and INS-GAS mice. RESULTS: H pylori induced a progressive loss of barrier function that was attenuated by inactivation of ureB, but not cagA, cagE, or vacA. Reductions in transepithelial electrical resistance were also dependent on functional urease activity. H pylori increased MLC phosphorylation in epithelial monolayers; this was significantly decreased by inhibition of MLCK or Rho kinase or by loss of UreB. H pylori infection of either cultured monolayers or hypergastrinemic INS-GAS mice induced occludin endocytosis, reflecting cytoskeletally mediated disruption of tight junctions. CONCLUSIONS: H pylori increases MLC phosphorylation, occludin internalization and barrier dysfunction in gastric epithelial cells. This process requires functional urease activity and is independent of the cag pathogenicity island or VacA. These data provide new insights into the mechanisms by which H pylori disrupts gastric barrier function.


Subject(s)
Gastric Mucosa/metabolism , Helicobacter pylori/pathogenicity , Myosin Type II/metabolism , Tight Junctions/physiology , Urease/physiology , Animals , Cells, Cultured , Electric Impedance , Gastric Mucosa/chemistry , Helicobacter pylori/enzymology , Membrane Proteins/analysis , Mice , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/metabolism , Occludin , Phosphorylation , rho-Associated Kinases/physiology
18.
FEMS Microbiol Lett ; 268(2): 135-43, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17313591

ABSTRACT

Helicobacter pylori is a Gram-negative microaerophilic organism that colonizes the gastric mucosa of humans. Helicobacter pylori is one of the most common infections in humans and results in the development of gastritis in all infected individuals, although the majority of people are asymptomatic. A subset of infected people develop serious disease including duodenal ulceration and gastric cancer. Helicobacter pylori exhibits many striking characteristics. It lives in the hostile environment of the stomach and displays a very strict host and tissue tropism. Despite a vigorous immune response, infection persists for the lifetime of the host unless eradicated with antimicrobials. Why H. pylori is so pathogenic in some individuals and not in others is unknown but is thought to be due to a variety of host, environmental and bacterial factors. In this review, some of the bacterial factors that mediate colonization of the gastric mucosa and play a role in the pathogenesis of this organism have been considered.


Subject(s)
Helicobacter Infections/physiopathology , Helicobacter pylori/physiology , Helicobacter pylori/pathogenicity , Adaptation, Physiological , Antigens, Bacterial/physiology , Bacterial Adhesion/physiology , Bacterial Proteins/physiology , Gastric Acid , Gastric Mucosa/microbiology , Humans , Urease/physiology , Virulence , Virulence Factors
20.
J Leukoc Biol ; 79(6): 1214-25, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16543403

ABSTRACT

Previous studies have demonstrated that Helicobacter pylori (Hp) delays its entry into macrophages and persists inside megasomes, which are poorly acidified and accumulate early endosome autoantigen 1. Herein, we explored the role of Hp urease in bacterial survival in murine peritoneal macrophages and J774 cells. Plasmid-free mutagenesis was used to replace ureA and ureB with chloramphenicol acetyltransferase in Hp Strains 11637 and 11916. ureAB null Hp lacked detectable urease activity and did not express UreA or UreB as judged by immunoblotting. Deletion of ureAB had no effect on Hp binding to macrophages or the rate or extent of phagocytosis. However, intracellular survival of mutant organisms was impaired significantly. Immunofluorescence microscopy demonstrated that (in contrast to parental organisms) mutant Hp resided in single phagosomes, which were acidic and accumulated the lysosome marker lysosome-associated membrane protein-1 but not early endosome autoantigen 1. A similar phenotype was observed for spontaneous urease mutants derived from Hp Strain 60190. Treatment of macrophages with bafilomycin A1, NH4Cl, or chloroquine prevented acidification of phagosomes containing mutant Hp. However, only ammonium chloride enhanced bacterial viability significantly. Rescue of ureAB null organisms was also achieved by surface adsorption of active urease. Altogether, our data indicate a role for urease and urease-derived ammonia in megasome formation and Hp survival.


Subject(s)
Helicobacter pylori/enzymology , Macrophages/microbiology , Phagosomes/physiology , Urease/physiology , Adsorption , Ammonia/metabolism , Ammonium Chloride/pharmacology , Animals , Autoantigens/analysis , Cell Line/microbiology , Chloroquine/pharmacology , Enzyme Induction , Female , Gene Deletion , Genes, Reporter , Helicobacter pylori/classification , Helicobacter pylori/genetics , Helicobacter pylori/physiology , Hydrogen-Ion Concentration , Lysosomal Membrane Proteins/analysis , Macrolides/pharmacology , Macrophages, Peritoneal/microbiology , Membrane Fusion , Membrane Proteins/analysis , Mice , Microscopy, Fluorescence , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Phagocytosis , Phagosomes/chemistry , Phagosomes/microbiology , Phenotype , Species Specificity , Urease/genetics , Urease/pharmacology , Vesicular Transport Proteins
SELECTION OF CITATIONS
SEARCH DETAIL