Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Microbiol Spectr ; 10(6): e0314222, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36377916

ABSTRACT

Ordered transposon libraries are a valuable resource for many bacterial species, especially those with difficult methods for generating targeted genetic mutations. Here, we present the construction of an ordered transposon library for the bacterial urinary tract pathogen Proteus mirabilis strain HI4320. This library will facilitate future studies into P. mirabilis biology. For large experimental screens, it may be used to overcome bottleneck constraints and avoid biased outcomes resulting from gene length. For smaller studies, the library allows sidestepping the laborious construction of single targeted mutants. This library, containing 18,432 wells, was condensed into a smaller library containing 1,728 mutants. Each selected mutant had a single transposon insertion in an open reading frame, covering 45% of predicted genes encoded by P. mirabilis HI4320. This coverage was lower than expected and was due both to library wells with no mapped insertions and a surprisingly high proportion of mixed clones and multiple transposon insertion events. We offer recommendations for improving future library construction and suggestions for how to use this P. mirabilis library resource. IMPORTANCE Ordered libraries facilitate large genetic screens by guaranteeing high genomic coverage with a minimal number of mutants, and they can save time and effort by reducing the need to construct targeted mutations. This resource is now available for P. mirabilis, a common and complicating agent of catheter-associated urinary tract infection. We also present obstacles encountered during library construction with the goal to aid others who would like to construct ordered transposon libraries in other species.


Subject(s)
Proteus Infections , Urinary Tract Infections , Urinary Tract , Humans , DNA Transposable Elements , Proteus mirabilis/genetics , Urinary Tract Infections/microbiology , Gene Library , Proteus Infections/genetics , Proteus Infections/microbiology
2.
Mol Microbiol ; 118(3): 125-144, 2022 09.
Article in English | MEDLINE | ID: mdl-35970717

ABSTRACT

Proteus mirabilis is a common cause of urinary tract infection, especially in catheterized individuals. Amino acids are the predominant nutrient for bacteria during growth in urine, and our prior studies identified several amino acid import and catabolism genes as fitness factors for P. mirabilis catheter-associated urinary tract infection (CAUTI), particularly those for d- and l-serine. In this study, we sought to determine the hierarchy of amino acid utilization by P. mirabilis and to examine the relative importance of d- vs l-serine catabolism for critical steps in CAUTI development and progression. Herein, we show that P. mirabilis preferentially catabolizes l-serine during growth in human urine, followed by d-serine, threonine, tyrosine, glutamine, tryptophan, and phenylalanine. Independently disrupting catabolism of either d- or l-serine has minimal impact on in vitro phenotypes while completely disrupting both pathways decreases motility, biofilm formation, and fitness due to perturbation of membrane potential and cell wall biosynthesis. In a mouse model of CAUTI, loss of either serine catabolism system decreased fitness, but disrupting l-serine catabolism caused a greater fitness defect than disrupting d-serine catabolism. We, therefore, conclude that the hierarchical utilization of amino acids may be a critical component of P. mirabilis colonization and pathogenesis within the urinary tract.


Subject(s)
Proteus Infections , Urinary Tract Infections , Animals , Catheters , Humans , Mice , Proteus Infections/genetics , Proteus Infections/microbiology , Proteus mirabilis/metabolism , Serine/metabolism , Urinary Tract Infections/microbiology , Urinary Tract Infections/pathology
3.
PLoS Pathog ; 15(6): e1007825, 2019 06.
Article in English | MEDLINE | ID: mdl-31220184

ABSTRACT

Medical devices, such as contact lenses, bring bacteria in direct contact with human cells. Consequences of these host-pathogen interactions include the alteration of mammalian cell surface architecture and induction of cellular death that renders tissues more susceptible to infection. Gram-negative bacteria known to induce cellular blebbing by mammalian cells, Pseudomonas and Vibrio species, do so through a type III secretion system-dependent mechanism. This study demonstrates that a subset of bacteria from the Enterobacteriaceae bacterial family induce cellular death and membrane blebs in a variety of cell types via a type V secretion-system dependent mechanism. Here, we report that ShlA-family cytolysins from Proteus mirabilis and Serratia marcescens were required to induce membrane blebbling and cell death. Blebbing and cellular death were blocked by an antioxidant and RIP-1 and MLKL inhibitors, implicating necroptosis in the observed phenotypes. Additional genetic studies determined that an IgaA family stress-response protein, GumB, was necessary to induce blebs. Data supported a model where GumB and shlBA are in a regulatory circuit through the Rcs stress response phosphorelay system required for bleb formation and pathogenesis in an invertebrate model of infection and proliferation in a phagocytic cell line. This study introduces GumB as a regulator of S. marcescens host-pathogen interactions and demonstrates a common type V secretion system-dependent mechanism by which bacteria elicit surface morphological changes on mammalian cells. This type V secretion-system mechanism likely contributes bacterial damage to the corneal epithelial layer, and enables access to deeper parts of the tissue that are more susceptible to infection.


Subject(s)
Bacterial Toxins/metabolism , Epithelial Cells/metabolism , Epithelium, Corneal/metabolism , Proteus Infections/metabolism , Proteus/metabolism , Serratia Infections/metabolism , Serratia marcescens/metabolism , Animals , Bacterial Toxins/genetics , Cell Death , Epithelial Cells/microbiology , Epithelial Cells/pathology , Epithelium, Corneal/microbiology , Epithelium, Corneal/pathology , Humans , Mice , Perforin/genetics , Perforin/metabolism , Proteus/genetics , Proteus Infections/genetics , Proteus Infections/microbiology , Proteus Infections/pathology , RAW 264.7 Cells , Serratia Infections/genetics , Serratia Infections/microbiology , Serratia Infections/pathology , Serratia marcescens/genetics , Swine , Type V Secretion Systems/genetics , Type V Secretion Systems/metabolism
4.
PLoS Pathog ; 15(4): e1007653, 2019 04.
Article in English | MEDLINE | ID: mdl-31009518

ABSTRACT

The Gram-negative bacterium Proteus mirabilis is a common cause of catheter-associated urinary tract infections (CAUTI), which can progress to secondary bacteremia. While numerous studies have investigated experimental infection with P. mirabilis in the urinary tract, little is known about pathogenesis in the bloodstream. This study identifies the genes that are important for survival in the bloodstream using a whole-genome transposon insertion-site sequencing (Tn-Seq) approach. A library of 50,000 transposon mutants was utilized to assess the relative contribution of each non-essential gene in the P. mirabilis HI4320 genome to fitness in the livers and spleens of mice at 24 hours following tail vein inoculation compared to growth in RPMI, heat-inactivated (HI) naïve serum, and HI acute phase serum. 138 genes were identified as ex vivo fitness factors in serum, which were primarily involved in amino acid transport and metabolism, and 143 genes were identified as infection-specific in vivo fitness factors for both spleen and liver colonization. Infection-specific fitness factors included genes involved in twin arginine translocation, ammonia incorporation, and polyamine biosynthesis. Mutants in sixteen genes were constructed to validate both the ex vivo and in vivo results of the transposon screen, and 12/16 (75%) exhibited the predicted phenotype. Our studies indicate a role for the twin arginine translocation (tatAC) system in motility, translocation of potential virulence factors, and fitness within the bloodstream. We also demonstrate the interplay between two nitrogen assimilation pathways in the bloodstream, providing evidence that the GS-GOGAT system may be preferentially utilized. Furthermore, we show that a dual-function arginine decarboxylase (speA) is important for fitness within the bloodstream due to its role in putrescine biosynthesis rather than its contribution to maintenance of membrane potential. This study therefore provides insight into pathways needed for fitness within the bloodstream, which may guide strategies to reduce bacteremia-associated mortality.


Subject(s)
Ammonia/metabolism , Arginine/metabolism , Bacteremia/microbiology , Polyamines/metabolism , Proteus Infections/microbiology , Proteus mirabilis/growth & development , Virulence Factors/metabolism , Animals , Bacteremia/genetics , Bacteremia/metabolism , DNA Transposable Elements , Female , Genetic Fitness , High-Throughput Nucleotide Sequencing , Mice , Mice, Inbred CBA , Phenotype , Proteus Infections/genetics , Proteus Infections/metabolism , Translocation, Genetic , Virulence Factors/genetics
6.
PLoS Pathog ; 13(6): e1006434, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28614382

ABSTRACT

The Gram-negative bacterium Proteus mirabilis is a leading cause of catheter-associated urinary tract infections (CAUTIs), which are often polymicrobial. Numerous prior studies have uncovered virulence factors for P. mirabilis pathogenicity in a murine model of ascending UTI, but little is known concerning pathogenesis during CAUTI or polymicrobial infection. In this study, we utilized five pools of 10,000 transposon mutants each and transposon insertion-site sequencing (Tn-Seq) to identify the full arsenal of P. mirabilis HI4320 fitness factors for single-species versus polymicrobial CAUTI with Providencia stuartii BE2467. 436 genes in the input pools lacked transposon insertions and were therefore concluded to be essential for P. mirabilis growth in rich medium. 629 genes were identified as P. mirabilis fitness factors during single-species CAUTI. Tn-Seq from coinfection with P. stuartii revealed 217/629 (35%) of the same genes as identified by single-species Tn-Seq, and 1353 additional factors that specifically contribute to colonization during coinfection. Mutants were constructed in eight genes of interest to validate the initial screen: 7/8 (88%) mutants exhibited the expected phenotypes for single-species CAUTI, and 3/3 (100%) validated the expected phenotypes for polymicrobial CAUTI. This approach provided validation of numerous previously described P. mirabilis fitness determinants from an ascending model of UTI, the discovery of novel fitness determinants specifically for CAUTI, and a stringent assessment of how polymicrobial infection influences fitness requirements. For instance, we describe a requirement for branched-chain amino acid biosynthesis by P. mirabilis during coinfection due to high-affinity import of leucine by P. stuartii. Further investigation of genes and pathways that provide a competitive advantage during both single-species and polymicrobial CAUTI will likely provide robust targets for therapeutic intervention to reduce P. mirabilis CAUTI incidence and severity.


Subject(s)
Catheter-Related Infections/microbiology , Coinfection/genetics , Proteus Infections/genetics , Proteus mirabilis/genetics , Proteus mirabilis/pathogenicity , Urinary Tract Infections/microbiology , Animals , DNA Transposable Elements , Disease Models, Animal , Genetic Fitness/genetics , Humans , Mice , Mice, Inbred CBA , Mutagenesis , Virulence Factors/genetics
7.
Proc Natl Acad Sci U S A ; 113(16): 4494-9, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27044107

ABSTRACT

The catheter-associated uropathogenProteus mirabilisfrequently causes urinary stones, but little has been known about the initial stages of bladder colonization and stone formation. We found thatP. mirabilisrapidly invades the bladder urothelium, but generally fails to establish an intracellular niche. Instead, it forms extracellular clusters in the bladder lumen, which form foci of mineral deposition consistent with development of urinary stones. These clusters elicit a robust neutrophil response, and we present evidence of neutrophil extracellular trap generation during experimental urinary tract infection. We identified two virulence factors required for cluster development: urease, which is required for urolithiasis, and mannose-resistantProteus-like fimbriae. The extracellular cluster formation byP. mirabilisstands in direct contrast to uropathogenicEscherichia coli, which readily formed intracellular bacterial communities but not luminal clusters or urinary stones. We propose that extracellular clusters are a key mechanism ofP. mirabilissurvival and virulence in the bladder.


Subject(s)
Bacterial Proteins , Fimbriae, Bacterial , Proteus Infections , Proteus mirabilis , Urease , Urinary Bladder Calculi , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Disease Models, Animal , Female , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Mice , Mice, Inbred CBA , Proteus Infections/genetics , Proteus Infections/metabolism , Proteus Infections/pathology , Proteus mirabilis/genetics , Proteus mirabilis/metabolism , Proteus mirabilis/pathogenicity , Urease/genetics , Urease/metabolism , Urinary Bladder/microbiology , Urinary Bladder/pathology , Urinary Bladder Calculi/genetics , Urinary Bladder Calculi/metabolism , Urinary Bladder Calculi/microbiology , Urinary Bladder Calculi/pathology , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/metabolism , Uropathogenic Escherichia coli/pathogenicity
8.
Immunity ; 42(4): 744-55, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25862092

ABSTRACT

The microbiota stimulates inflammation, but the signaling pathways and the members of the microbiota involved remain poorly understood. We found that the microbiota induces interleukin-1ß (IL-1ß) release upon intestinal injury and that this is mediated via the NLRP3 inflammasome. Enterobacteriaceae and in particular the pathobiont Proteus mirabilis, induced robust IL-1ß release that was comparable to that induced by the pathogen Salmonella. Upon epithelial injury, production of IL-1ß in the intestine was largely mediated by intestinal Ly6C(high) monocytes, required chemokine receptor CCR2 and was abolished by deletion of IL-1ß in CCR2(+) blood monocytes. Furthermore, colonization with P. mirabilis promoted intestinal inflammation upon intestinal injury via the production of hemolysin, which required NLRP3 and IL-1 receptor signaling in vivo. Thus, upon intestinal injury, selective members of the microbiota stimulate newly recruited monocytes to induce NLRP3-dependent IL-1ß release, which promotes inflammation in the intestine.


Subject(s)
Carrier Proteins/immunology , Inflammasomes/immunology , Interleukin-1beta/immunology , Microbiota/immunology , Monocytes/immunology , Symbiosis/immunology , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Carrier Proteins/genetics , Gene Expression Regulation , Hemolysin Proteins/genetics , Hemolysin Proteins/immunology , Inflammasomes/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Interleukin-1beta/genetics , Intestines/immunology , Intestines/injuries , Intestines/microbiology , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/microbiology , Monocytes/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , Proteus Infections/genetics , Proteus Infections/immunology , Proteus Infections/microbiology , Proteus Infections/pathology , Proteus mirabilis/immunology , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Salmonella/immunology , Salmonella Infections/genetics , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella Infections/pathology , Signal Transduction
9.
J Biol Chem ; 288(22): 15668-76, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23572531

ABSTRACT

Previously, we reported that the speA gene, encoding arginine decarboxylase, is required for swarming in the urinary tract pathogen Proteus mirabilis. In addition, this previous study suggested that putrescine may act as a cell-to-cell signaling molecule (Sturgill, G., and Rather, P. N. (2004) Mol. Microbiol. 51, 437-446). In this new study, PlaP, a putative putrescine importer, was characterized in P. mirabilis. In a wild-type background, a plaP null mutation resulted in a modest swarming defect and slightly decreased levels of intracellular putrescine. In a P. mirabilis speA mutant with greatly reduced levels of intracellular putrescine, plaP was required for the putrescine-dependent rescue of swarming motility. When a speA/plaP double mutant was grown in the presence of extracellular putrescine, the intracellular levels of putrescine were greatly reduced compared with the speA mutant alone, indicating that PlaP functioned as the primary putrescine importer. In urothelial cell invasion assays, a speA mutant exhibited a 50% reduction in invasion when compared with wild type, and this defect could be restored by putrescine in a PlaP-dependent manner. The putrescine analog Triamide-44 partially inhibited the uptake of putrescine by PlaP and decreased both putrescine stimulated swarming and urothelial cell invasion in a speA mutant.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Proteus Infections/metabolism , Proteus mirabilis/metabolism , Putrescine/metabolism , Urothelium/microbiology , Bacterial Proteins/genetics , Carrier Proteins/genetics , Cell Line , Humans , Mutation , Proteus Infections/genetics , Proteus Infections/microbiology , Proteus mirabilis/genetics , Proteus mirabilis/pathogenicity , Urothelium/pathology
10.
Mol Biol Rep ; 40(4): 3429-35, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23283741

ABSTRACT

Escherichia coli and Proteus mirabilis are important urinary tract pathogens. The constant increase in the antibiotic resistance of clinical bacterial strains has become an important clinical problem. The aim of this study was to compare the antibiotic resistance of 141 clinical (Sweden and Poland) and 42 laboratory (Czech Republic) P. mirabilis strains and 129 clinical (Poland) uropathogenic E. coli strains. The proportion of unique versus diverse patterns in Swedish clinical and laboratory P. mirabilis strain collections was comparable. Notably, a similar proportion of unique versus diverse patterns was observed in Polish clinical P. mirabilis and E. coli strain collections. Mathematical models of the antibiotic resistance of E. coli and P. mirabilis strains based on Kohonen networks and association analysis are presented. In contrast to the three clinical strain collections, which revealed complex associations with the antibiotics tested, laboratory P. mirabilis strains provided simple antibiotic association diagrams. The monitoring of antibiotic resistance patterns of clinical E. coli and P. mirabilis strains plays an important role in the treatment procedures for urinary tract infections and is important in the context of the spreading drug resistance in uropathogenic strain populations. The adaptability and flexibility of the genomes of E. coli and P. mirabilis strains are discussed.


Subject(s)
Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Proteus mirabilis/genetics , Urinary Tract Infections/microbiology , Czech Republic , Escherichia coli/pathogenicity , Genome, Bacterial , Humans , Poland , Proteus Infections/drug therapy , Proteus Infections/genetics , Proteus Infections/microbiology , Proteus mirabilis/pathogenicity , Sweden , Urinary Tract Infections/drug therapy , Urinary Tract Infections/genetics
11.
Am J Clin Pathol ; 137(4): 620-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22431539

ABSTRACT

In the present study, nonduplicate, clinical isolates of extended-spectrum ß-lactamase (ESBL)-producing Escherichia coli, Klebsiella spp, and Proteus mirabilis were collected during a 10-year period from 2000 to 2009 at several hospitals in the Kinki region, Japan. The detection rate of E coli markedly increased from 0.24% to 7.25%. The detection rate of Klebsiella pneumoniae increased from 0% to 2.44% and that of P mirabilis from 6.97% to 12.85%. The most frequently detected genotypes were the CTX-M9 group for E coli, the CTX-M2 group for K pneumoniae, and the CTX-M2 group for P mirabilis. E coli clone O25:H4-ST131 producing CTX-M-15, which is spreading worldwide, was first detected in 2007. The most common replicon type of E coli was the IncF type, particularly FIB, detected in 466 strains (69.7%). Of the K pneumoniae strains, 47 (55.3%) were of the IncN type; 77 P mirabilis strains (96.3%) were of the IncT type. In the future, the surveillance of various resistant bacteria, mainly ESBL-producing Enterobacteriaceae, should be expanded to prevent their spread.


Subject(s)
Escherichia coli Infections/epidemiology , Escherichia coli/genetics , Klebsiella Infections/epidemiology , Klebsiella/genetics , Proteus Infections/epidemiology , Proteus mirabilis/genetics , beta-Lactamases/genetics , Bacterial Typing Techniques , DNA, Bacterial/genetics , Escherichia coli/isolation & purification , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Humans , Japan/epidemiology , Klebsiella/isolation & purification , Klebsiella Infections/genetics , Klebsiella Infections/microbiology , Molecular Epidemiology , Proteus Infections/genetics , Proteus Infections/microbiology , Proteus mirabilis/isolation & purification
12.
Front Biosci (Elite Ed) ; 1(2): 577-86, 2009 06 01.
Article in English | MEDLINE | ID: mdl-19482674

ABSTRACT

Rheumatoid arthritis is a crippling and disabling joint disease affecting over 20 million people. It occurs predominantly in women and smokers, and affects the HLA-DR1/4 individuals who carry the "shared epitope" of amino acids EQRRAA. The cause of this disease was investigated by the methods of the philosopher of science Karl Popper who suggested that scientific research should be based on bold conjectures and critical refutations. The "Popper sequences" generate new facts which then change or alter the original problem. The new facts must then be explained by any new theory. Using the "molecular mimicry" model, it was found that Proteus bacteria possess an amino acid sequence ESRRAL in haemolysin which resembles the, shared epitope, and another sequence in urease which resembles type XI collagen. Antibodies to Proteus bacteria have been found in 14 different countries. It would appear that rheumatoid arthritis is caused by an upper urinary tract infection by Proteus bacteria. Anti-Proteus therapy should be assessed in the management of this disease separately or in conjunction with existing modalities of therapy.


Subject(s)
Amino Acid Motifs/genetics , Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/microbiology , HLA-DR1 Antigen/genetics , Molecular Mimicry/genetics , Proteus Infections/complications , Proteus/genetics , Urinary Tract Infections/complications , Antibodies, Bacterial/genetics , Collagen Type XI/immunology , Female , Hemolysin Proteins/genetics , Humans , Proteus Infections/genetics , Risk Factors , Smoking/adverse effects , Urease/genetics , Urinary Tract Infections/microbiology
13.
J Urol ; 181(5): 2332-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19303096

ABSTRACT

PURPOSE: Proteus mirabilis is a common cause of urinary tract infection. We determined the role of Tamm-Horsfall protein as a host defense factor against the cystitis and pyelonephritis caused by P. mirabilis. MATERIALS AND METHODS: We generated Tamm-Horsfall protein gene knockout mice using homologous recombination. We introduced P. mirabilis transurethrally into the bladder of Tamm-Horsfall protein deficient (THP(-/-)) and genetically similar WT (THP(+/+)) mice. We cultured urine to quantitate the degree of bacteriuria. We examined bladders and kidneys grossly and histomorphometrically to determine the intensity of inflammation. RESULTS: THP(-/-) mice had more severe bacteriuria and cystitis than THP(+/+) mice. THP(-/-) mice had more pyelonephritic abscesses than THP(+/+) mice. The severity of histological pyelonephritis on semiquantitative histomorphometric analysis appeared to be greater in THP(-/-) mice. The difference between the 2 groups approached but did not attain statistical significance (p = 0.053). CONCLUSION: Tamm-Horsfall protein acts as a host defense factor against P. mirabilis induced urinary tract infection.


Subject(s)
Cystitis/pathology , Cystitis/prevention & control , Mucoproteins/genetics , Proteus Infections/prevention & control , Animals , Bacteriuria/genetics , Bacteriuria/physiopathology , Biopsy, Needle , Cystitis/genetics , Disease Models, Animal , Genetic Markers , Genetic Predisposition to Disease , Immunohistochemistry , Mice , Mice, Knockout , Mucoproteins/metabolism , Probability , Proteus Infections/genetics , Proteus Infections/physiopathology , Proteus mirabilis/pathogenicity , Random Allocation , Sensitivity and Specificity , Statistics, Nonparametric , Urinary Tract Infections/genetics , Urinary Tract Infections/pathology , Urinary Tract Infections/prevention & control , Uromodulin
14.
Infect Immun ; 76(11): 4859-64, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18725420

ABSTRACT

Our knowledge of pathogenesis has benefited from a better understanding of the roles of specific virulence factors in disease. To determine the role of the virulence factor ZapA, a 54-kDa metalloproteinase of Proteus mirabilis, in prostatitis, rats were infected with either wild-type (WT) P. mirabilis or its isogenic ZapA(-) mutant KW360. The WT produced both acute and chronic prostatitis showing the typical histological progressions that are the hallmarks of these diseases. Infection with the ZapA(-) mutant, however, resulted in reduced levels of acute prostatitis, as determined from lower levels of tissue damage, bacterial colonization, and inflammation. Further, the ZapA(-) mutant failed to establish a chronic infection, in that bacteria were cleared from the prostate, inflammation was resolved, and tissue was seen to be healing. Clearance from the prostate was not the result of a reduced capacity of the ZapA(-) mutant to form biofilms in vitro. These finding clearly define ZapA as an important virulence factor in both acute and chronic bacterial prostatitis.


Subject(s)
Bacterial Proteins/metabolism , Prostatitis/metabolism , Proteus Infections/metabolism , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Chronic Disease , Disease Models, Animal , Male , Prostatitis/genetics , Prostatitis/pathology , Proteus Infections/genetics , Proteus Infections/pathology , Proteus mirabilis/genetics , Proteus mirabilis/metabolism , Proteus mirabilis/pathogenicity , Rats , Rats, Sprague-Dawley , Virulence Factors/genetics
15.
Scand J Rheumatol ; 32(1): 2-11, 2003.
Article in English | MEDLINE | ID: mdl-12635939

ABSTRACT

Rheumatoid arthritis (RA) is a chronic disease, affecting women more than men, especially in those possessing the "shared epitope" (EQK/RRAA) amino acid sequences present in HLA-DR1/4 molecules. Proteus mirabilis carries sequences showing molecular mimicry to the "shared epitope" and to type XI collagen of hyaline cartilage. Elevated levels of antibodies to P. mirabilis have been reported from 14 different countries involving 1375 RA patients and the microbe has been isolated from urine cultures of such patients. Our working hypothesis is that the disease develops as a result of repeated episodes of Proteus upper urinary tract infections. Prospective studies involving the trial of anti-Proteus measures in RA patients should be evaluated in the management of this disease. Antibiotics, high fluid intake, and fruit extracts, such as cranberry juice, have all been found to be effective in the treatment of urinary tract infections. Such measures could be used as possible additional adjuncts to the standard therapy with NSAIDs and DMARDs.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Arthritis, Rheumatoid , Proteus Infections/complications , Proteus Infections/drug therapy , Proteus mirabilis/pathogenicity , Urinary Tract Infections/drug therapy , Urinary Tract Infections/etiology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/etiology , Female , Genetic Predisposition to Disease , Humans , Male , Proteus Infections/genetics , Proteus mirabilis/genetics , Proteus mirabilis/immunology , Time Factors
16.
Infect Immun ; 66(1): 330-5, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9423875

ABSTRACT

Proteus mirabilis, a cause of complicated urinary tract infection, expresses urease when exposed to urea. While it is recognized that the positive transcriptional activator UreR induces gene expression, the levels of expression of the enzyme during experimental infection are not known. To investigate in vivo expression of P. mirabilis urease, the gene encoding green fluorescent protein (GFP) was used to construct reporter fusions. Translational fusions of urease accessory gene ureD, which is preceded by a urea-inducible promoter, were made with gfp (modified to express S65T/V68L/S72A [B. P. Cormack et al. Gene 173:33-38, 1996]). Constructs were confirmed by sequencing of the fusion junctions. UreD-GFP fusion protein was induced by urea in both Escherichia coli DH5alpha and P. mirabilis HI4320. By using Western blotting with antiserum raised against GFP, expression level was shown to correlate with urea concentration (tested from 0 to 500 mM), with highest induction at 200 to 500 mM urea. Fluorescent E. coli and P. mirabilis bacteria were observed by fluorescence microscopy following urea induction, and the fluorescence intensity of GFP in cell lysates was measured by spectrophotofluorimetry. P. mirabilis HI4320 carrying the UreD-GFP fusion plasmid was transurethrally inoculated into the bladders of CBA mice. One week postchallenge, fluorescent bacteria were detected in thin sections of both bladder and kidney samples; the fluorescence intensity of bacteria in bladder tissue was higher than that in the kidney. Kidneys were primarily infected with single-cell-form fluorescent bacteria, while aggregated bacterial clusters were observed in the bladder. Elongated swarmer cells were only rarely observed. These observations demonstrate that urease is expressed in vivo and that using GFP as a reporter protein is a viable approach to investigate in vivo expression of P. mirabilis virulence genes in experimental urinary tract infection.


Subject(s)
Gene Expression , Luminescent Proteins/genetics , Proteus Infections/genetics , Proteus mirabilis/genetics , Urease/genetics , Urinary Tract Infections/microbiology , Animals , Artificial Gene Fusion , Bacterial Proteins/genetics , Blotting, Western , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Genes, Reporter , Green Fluorescent Proteins , Kidney/microbiology , Luminescent Proteins/immunology , Luminescent Proteins/metabolism , Mice , Mice, Inbred CBA , Microscopy, Fluorescence , Plasmids , Polymerase Chain Reaction , Promoter Regions, Genetic , Proteus mirabilis/metabolism , Recombination, Genetic , Sequence Analysis, DNA , Spectrophotometry , Urea/pharmacology , Urease/metabolism , Urinary Bladder/microbiology , Urinary Tract Infections/genetics
17.
Mol Microbiol ; 23(5): 1009-19, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9076737

ABSTRACT

Proteus mirabilis, associated with complicated urinary tract infection, expresses mannose-resistant/Proteus-like (MR/P) fimbriae. Expression of these surface structures, which mediate haemagglutination and have a demonstrated role in virulence, undergoes phase variation. By DNA sequence analysis, a 252 bp invertible element was found in the intergenic region between mrpl, the putative site-specific recombinase gene, and mrpA, the primary structural subunit gene. The invertible segment is flanked by identical 21 bp inverted repeats and the presumptive half-sites for recombinase binding show homology to those recognized by FimB and FimE encoded by the Escherichia coli fim (Type 1 fimbriae) gene cluster. When amplified by the polymerase chain reaction (PCR) from static broth cultures expressing MR/P fimbriae, the switch region was found in both ON and OFF positions. When PCR was used to amplify agar cultures which do not express the fimbriae, the switch region was OFF only. A canonical sigma 70 promoter inside the invertible element drives the transcription of mrpA when in the ON position; in the OFF position it is directed away from mrpA but does not appear to drive expression of mrpI. The mrpI gene was able to confer inversion of the mrp switch region in trans from both ON to OFF and OFF to ON. To examine the position of the switch in vivo, urine, bladder, and kidneys from mice transurethrally infected with P. mirabilis were used to prepare template DNA for PCR amplification. In the absence of urolithiasis (urease-mediated stone formation), the switch was found 100% in the ON position, a condition never observed following in vitro culture. We conclude that MR/P phase variation is regulated at the transcriptional level by the action of MrpI on an invertible element and that there is strong selective pressure for the expression of MR/P fimbriae in vivo.


Subject(s)
Escherichia coli Proteins , Fimbriae, Bacterial/genetics , Gene Expression Regulation, Bacterial , Integrases , Mannose/metabolism , Proteus Infections/genetics , Proteus Infections/microbiology , Proteus mirabilis/genetics , Urinary Tract Infections/genetics , Urinary Tract Infections/microbiology , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Base Sequence , Blotting, Southern , Cloning, Molecular , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/physiology , DNA, Bacterial/analysis , DNA, Bacterial/genetics , DNA-Binding Proteins/genetics , Escherichia coli/genetics , Female , Genetic Complementation Test , Kidney/microbiology , Lac Operon , Mice , Mice, Inbred CBA , Microscopy, Immunoelectron , Molecular Sequence Data , Plasmids , Polymerase Chain Reaction , Promoter Regions, Genetic , Proteus Infections/urine , Proteus mirabilis/pathogenicity , Recombinases , Recombination, Genetic , Repetitive Sequences, Nucleic Acid , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sigma Factor/genetics , Transcription, Genetic , Urinary Bladder/microbiology , Urinary Tract Infections/urine , Virulence , beta-Galactosidase/metabolism
18.
Infect Immun ; 64(12): 5332-40, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8945585

ABSTRACT

To examine the role of flagella in pathogenesis of urinary tract infection caused by Proteus mirabilis, we constructed a nonmotile, nonswarming flagellum mutant of strain WPM111 (an hpmA hemolysin mutant of strain BA6163, chosen because of its lack of in vitro cytotoxicity in renal epithelial cell internalization studies). A nonpolar mutation was introduced into the flaD gene, which encodes the flagellar cap protein. This mutation does not affect the synthesis of flagellin but rather prevents the assembly of an intact flagellar filament. In in vitro assays, the genetically characterized nonmotile mutant was found to be internalized by cultured human renal proximal tubular epithelial cells in numbers less than 1% of those of the flagellated parent strain. Internalization of the nonmotile mutant was increased significantly (14- to 21-fold) by centrifugation onto the monolayer. To assess virulence in vivo, CBA mice were challenged transurethrally with 10(7) CFU of P. mirabilis BA6163 (wild type) (n = 16), WPM111 (hpmA mutant) (n = 46), or BB2401 (hmpA flaD mutant) (n = 46). Differences in quantitative cultures between the parent strain and the hemolysin-negative mutant were not significant. However, the hpmA flaD mutant was recovered in numbers approximately 100-fold lower than those of the hmpA mutant or the wild-type parent strain and thus was clearly attenuated. We conclude that while hemolysin does not significantly influence virulence, flagella contribute significantly to the ability of P. mirabilis to colonize the urinary tract and cause acute pyelonephritis in an experimental model of ascending urinary tract infection.


Subject(s)
Flagella/genetics , Kidney/microbiology , Proteus Infections/genetics , Proteus mirabilis/genetics , Urinary Tract Infections/microbiology , Animals , Cells, Cultured , Epithelium/microbiology , Humans , Mice , Mice, Inbred CBA , Mutation , Proteus Infections/microbiology , Proteus mirabilis/pathogenicity , Virulence/genetics
19.
Article in Russian | MEDLINE | ID: mdl-8067124

ABSTRACT

A total of 286 convalescents after toxico-infections caused by opportunistic microorganisms were examined. The panel of anti-HLA sera permitted the typing of 34 HLA antigens, class 1. As a result, the development of the disease was found to be directly associated with antigen HLA-B17 and inversely with antigen HLA-A2. The relationship of antigens of the HLA system with alimentary toxico-infections caused by opportunistic microorganisms manifested not by the absolute resistance or predisposition to the disease, but by an increased severity of the pathological process, paralleled by a decreased level of protective antigens and an increased one of provoking antigens of the HLA system.


Subject(s)
Foodborne Diseases/immunology , HLA Antigens/blood , Klebsiella Infections/immunology , Opportunistic Infections/immunology , Proteus Infections/immunology , Pseudomonas Infections/immunology , Adolescent , Adult , Chi-Square Distribution , Child , Disease Susceptibility , Female , Foodborne Diseases/epidemiology , Foodborne Diseases/genetics , Gene Frequency , HLA Antigens/genetics , Humans , Klebsiella Infections/epidemiology , Klebsiella Infections/genetics , Male , Middle Aged , Opportunistic Infections/epidemiology , Opportunistic Infections/genetics , Proteus Infections/epidemiology , Proteus Infections/genetics , Pseudomonas Infections/epidemiology , Pseudomonas Infections/genetics , Risk , Ukraine/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...