Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Antimicrob Chemother ; 78(2): 373-379, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36480295

ABSTRACT

BACKGROUND: Nitrofurantoin has been re-introduced as a first-choice antibiotic to treat uncomplicated acute urinary tract infections in England and Wales. Highly effective against common uropathogens such as Escherichia coli, its use is accompanied by a low incidence (<10%) of antimicrobial resistance. Resistance to nitrofurantoin is predominantly via the acquisition of loss-of-function, step-wise mutations in the nitroreductase genes nfsA and nfsB. OBJECTIVE: To explore the in situ evolution of NitR in E. coli isolates from 17 patients participating in AnTIC, a 12-month open label randomized controlled trial assessing the efficacy of antibiotic prophylaxis in reducing urinary tract infections (UTIs) incidence in clean intermittent self-catheterizing patients. METHODS: The investigation of NitR evolution in E. coli used general microbiology techniques and genetics to model known NitR mutations in NitSE. coli strains. RESULTS: Growth rate analysis identified a 2%-10% slower doubling time for nitrofurantoin resistant strains: NitS: 20.8Ć¢Ā€ĀŠĀ±Ć¢Ā€ĀŠ0.7 min compared to NitR: 23Ć¢Ā€ĀŠĀ±Ć¢Ā€ĀŠ0.8 min. Statistically, these data indicated no fitness advantage of evolved strains compared to the sensitive predecessor (P-valueĆ¢Ā€ĀŠ=Ć¢Ā€ĀŠ0.13). Genetic manipulation of E. coli to mimic NitR evolution, supported no fitness advantage (P-valueĆ¢Ā€ĀŠ=Ć¢Ā€ĀŠ0.22). In contrast, data argued that a first-step mutant gained a selective advantage, at sub-MIC (4-8 mg/L) nitrofurantoin concentrations. CONCLUSION: Correlation of these findings to nitrofurantoin pharmacokinetic data suggests that the low incidence of E. coli NitR, within the community, is driven by urine-based nitrofurantoin concentrations that selectively inhibit the growth of E. coli strains carrying the key first-step loss-of-function mutation.


Subject(s)
Escherichia coli Infections , Urinary Tract Infections , Uropathogenic Escherichia coli , Humans , Nitrofurantoin/pharmacology , Nitrofurantoin/therapeutic use , Uropathogenic Escherichia coli/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Urinary Tract Infections/microbiology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Microbial Sensitivity Tests
2.
J Bacteriol ; 202(13)2020 06 09.
Article in English | MEDLINE | ID: mdl-32312744

ABSTRACT

Flagellar gene expression is bimodal in Salmonella enterica Under certain growth conditions, some cells express the flagellar genes whereas others do not. This results in mixed populations of motile and nonmotile cells. In the present study, we found that two independent mechanisms control bimodal expression of the flagellar genes. One was previously found to result from a double negative-feedback loop involving the flagellar regulators RflP and FliZ. This feedback loop governs bimodal expression of class 2 genes. In this work, a second mechanism was found to govern bimodal expression of class 3 genes. In particular, class 3 gene expression is still bimodal, even when class 2 gene expression is not. Using a combination of experimental and modeling approaches, we found that class 3 bimodality results from the σ28-FlgM developmental checkpoint.IMPORTANCE Many bacterial use flagella to swim in liquids and swarm over surface. In Salmonella enterica, over 50 genes are required to assemble flagella. The expression of these genes is tightly regulated. Previous studies have found that flagellar gene expression is bimodal in S. enterica, which means that only a fraction of cells express flagellar genes and are motile. In the present study, we found that two separate mechanisms induce this bimodal response. One mechanism, which was previously identified, tunes the fraction of motile cells in response to nutrients. The other results from a developmental checkpoint that couples flagellar gene expression to flagellar assembly. Collectively, these results further our understanding of how flagellar gene expression is regulated in S. enterica.


Subject(s)
Bacterial Proteins/metabolism , Flagella/metabolism , Gene Expression Regulation, Bacterial , Salmonella enterica/metabolism , Bacterial Proteins/genetics , Flagella/genetics , Promoter Regions, Genetic , Salmonella enterica/genetics , Salmonella enterica/growth & development
3.
J Surg Res ; 235: 288-297, 2019 03.
Article in English | MEDLINE | ID: mdl-30691808

ABSTRACT

BACKGROUND: Pancreas transplantation restores insulin secretion in type 1 diabetes mellitus. The graft also produces exocrine secretions that can be drained enterically (enteric drainage [ED]) or via the bladder (bladder drainage [BD]). We suggest that in BD transplants, such secretions destroy bladder innate immunity, specifically host defense peptides/proteins (HDPs), which increases patient susceptibility to recurrent urinary tract infections (rUTIs). MATERIALS AND METHODS: BD and ED patient records were reviewed retrospectively for UTIs. Urine samples from ED and BD transplant recipients were analyzed for pH, the HDPs Ɵ-defensin 2 (HBD2) and lipocalin-2, and amylase concentrations. InĀ vitro, bacterial growth curves and antimicrobial assays were used to evaluate the effects of pH, HBD2, and HBD2Ā +Ā pancreatic digestive enzymes (pancreatin) on uropathogenic Escherichia coli (UPEC) survival and growth. RESULTS: Urinalysis revealed a significant difference in pH between the BD and ED cohorts (7.2Ā Ā±Ā 0.8 versus 6.7Ā Ā±Ā 0.8; PĀ =Ā 0.012). Urinary HDPs were measured and BD, but not ED, lipocalin-2 concentrations were significantly decreased compared with those of diabetics awaiting transplant (PĀ <Ā 0.05). InĀ vitro, an alkaline environment, pH 8.0, concomitant with the urine of the patient who underwent BD transplantation, significantly reduced UPEC growth (PĀ <Ā 0.05); addition of pancreatin to the growth medium was associated with a significant increase (PĀ <Ā 0.001) in growth rate. Antimicrobial data suggested significant UPEC killing in the presence of HBD2 (PĀ <Ā 0.01), but not in the presence of HBD2 + pancreatin (>12,500 amylase units). CONCLUSIONS: These inĀ vivo and inĀ vitro data suggest that BD pancreatic exocrine secretions inactivate the bladder innate defenses, which facilitate UPEC growth and underpins the increased susceptibility of patients who underwent BD pancreas transplantation to rUTIs.


Subject(s)
Pancreas Transplantation/adverse effects , Urinary Tract Infections/immunology , Adult , Cell Line , Female , Humans , Immunity, Innate , Male , Middle Aged , Pancreas Transplantation/methods , Pancreatin , Retrospective Studies , United Kingdom/epidemiology , Urinary Bladder/immunology , Urinary Tract Infections/epidemiology , Urine/chemistry , beta-Defensins/physiology
4.
Immun Ageing ; 16: 16, 2019.
Article in English | MEDLINE | ID: mdl-31338112

ABSTRACT

BACKGROUND: Age is a significant risk factor for recurrent urinary tract (rUTI) infections, but the clinical picture is often confused in older patients who also present with asymptomatic bacteriuria (ASB). Yet, how bacteriuria establishes in such patients and the factors underpinning and/or driving symptomatic UTI episodes are still not understood. To explore this further a pilot study was completed in which 30 male and female community based older patients (mean age 75y) presenting clinically with ASB / rUTIs and 15 control volunteers (72y) were recruited and monitored for up to 6 months. During this period symptomatic UTI episodes were recorded and urines collected for urinary cytokine and uropathogenic Escherichia coli (UPEC) analyses. RESULTS: Eighty-six per cent of patients carried E. coli (102Ā ≥ 105 CFU/ml urine) at some point throughout the study and molecular typing identified 26 different E. coli strains in total. Analyses of urine samples for ten different cytokines identified substantial patient variability. However, when examined longitudinally the pro-inflammatory markers, IL-1 and IL-8, and the anti-inflammatory markers, IL-5 and IL-10, were significantly different in the patient urines compared to those of the controls (PĀ < 0.0001). Furthermore, analysing the cytokine data of the rUTI susceptible cohort in relation to E. coli carriage, showed the mean IL-10 concentration to be significantly elevated (PĀ = 0.04), in patients displaying E. coli numbers ≥105 CFU/ml. CONCLUSIONS: These pilot study data suggest that bacteriuria, characteristic of older rUTI patients, is associated with an immune homeostasis in the urinary tract involving the synthesis and activities of the pro and anti-inflammatory cytokines IL-1, IL-5, IL-8 and IL-10. Data also suggests a role for IL-10 in regulating bacterial persistence.

5.
PLoS Pathog ; 12(3): e1005495, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26943926

ABSTRACT

The bacterial flagellar type III export apparatus utilizes ATP and proton motive force (PMF) to transport flagellar proteins to the distal end of the growing flagellar structure for self-assembly. The transmembrane export gate complex is a H+-protein antiporter, of which activity is greatly augmented by an associated cytoplasmic ATPase complex. Here, we report that the export gate complex can use sodium motive force (SMF) in addition to PMF across the cytoplasmic membrane to drive protein export. Protein export was considerably reduced in the absence of the ATPase complex and a pH gradient across the membrane, but Na+ increased it dramatically. Phenamil, a blocker of Na+ translocation, inhibited protein export. Overexpression of FlhA increased the intracellular Na+ concentration in the presence of 100 mM NaCl but not in its absence, suggesting that FlhA acts as a Na+ channel. In wild-type cells, however, neither Na+ nor phenamil affected protein export, indicating that the Na+ channel activity of FlhA is suppressed by the ATPase complex. We propose that the export gate by itself is a dual fuel engine that uses both PMF and SMF for protein export and that the ATPase complex switches this dual fuel engine into a PMF-driven export machinery to become much more robust against environmental changes in external pH and Na+ concentration.


Subject(s)
Flagella/metabolism , Proton-Translocating ATPases/metabolism , Salmonella/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Flagella/genetics , Hydrogen/metabolism , Mutation , Promoter Regions, Genetic/genetics , Protein Transport , Proton-Translocating ATPases/genetics , Salmonella/genetics , Sodium/metabolism
6.
J Bacteriol ; 196(12): 2216-26, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24706744

ABSTRACT

The assembly of the bacterial flagellum is exquisitely controlled. Flagellar biosynthesis is underpinned by a specialized type III secretion system that allows export of proteins from the cytoplasm to the nascent structure. Bacillus subtilis regulates flagellar assembly using both conserved and species-specific mechanisms. Here, we show that YvyG is essential for flagellar filament assembly. We define YvyG as an orthologue of the Salmonella enterica serovar Typhimurium type III secretion system chaperone, FlgN, which is required for the export of the hook-filament junction proteins, FlgK and FlgL. Deletion of flgN (yvyG) results in a nonmotile phenotype that is attributable to a decrease in hag translation and a complete lack of filament polymerization. Analyses indicate that a flgK-flgL double mutant strain phenocopies deletion of flgN and that overexpression of flgK-flgL cannot complement the motility defect of a ΔflgN strain. Furthermore, in contrast to previous work suggesting that phosphorylation of FlgN alters its subcellular localization, we show that mutation of the identified tyrosine and arginine FlgN phosphorylation sites has no effect on motility. These data emphasize that flagellar biosynthesis is differentially regulated in B. subtilis from classically studied Gram-negative flagellar systems and questions the biological relevance of some posttranslational modifications identified by global proteomic approaches.


Subject(s)
Bacillus subtilis/metabolism , Bacterial Proteins/metabolism , Flagella/physiology , Amino Acid Sequence , Bacillus subtilis/cytology , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial/physiology , Molecular Sequence Data , Movement , Mutation
7.
Mol Microbiol ; 83(4): 775-88, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22233518

ABSTRACT

FlgN chaperone acts as a bodyguard to protect its cognate substrates, FlgK and FlgL, from proteolysis in the cytoplasm. Docking of the FlgN-FlgK complex with the FliI ATPase of the flagellar type III export apparatus is key to the protein export process. However, a ΔfliH-fliI flhB(P28T) mutant forms some flagella even in the absence of FliH and FliI, raising the question of how FlgN promotes the export of its cognate substrates. Here, we report that the interaction of FlgN with an integral membrane export protein, FlhA, is directly involved in efficient protein export. A ΔfliH-fliI flhB(P28T) ΔflgN mutant caused extragenic suppressor mutations in the C-terminal domain of FlhA (FlhA(C) ). Pull-down assays using GST affinity chromatography showed an interaction between FlgN and FlhA(C) . The FlgN-FlgK complex bound to FlhA(C) and FliJ to form the FlgN-FlgK-FliJ-FlhA(C) complex. The FlgN-FlhA(C) interaction was enhanced by FlgK but not by FliJ. FlgN120 missing the last 20 residues still bound to FlgK and FliJ but not to FlhA(C) . A highly conserved Tyr-122 residue was required for the interaction with FlhA(C) . These results suggest that FlgN efficiently transfers FlgK/L subunits to FlhA(C) to promote their export.


Subject(s)
Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Protein Interaction Mapping , Bacterial Proteins/genetics , Gene Deletion , Membrane Proteins/genetics , Models, Biological , Models, Molecular , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Protein Transport , Suppression, Genetic
8.
Front Microbiol ; 14: 1093922, 2023.
Article in English | MEDLINE | ID: mdl-37032848

ABSTRACT

Uropathogenic Escherichia coli is a major cause of urinary tract infections. Analysis of the innate immune response in immortalized urothelial cells suggests that the bacterial flagellar subunit, flagellin, is key in inducing host defenses. A panel of 48 clinical uro-associated E. coli isolates recovered from either cystitis, pyelonephritis asymptomatic bacteriuria (ABU) or UTI-associated bacteraemia infections were characterized for motility and their ability to induce an innate response in urothelial cells stably transfected with a NF-κB luciferase reporter. Thirty-two isolates (67%) were identified as motile with strains recovered from cystitis patients exhibiting an uneven motility distribution pattern; seven of the cystitis isolates were associated with a > 5-fold increase in NF-κB signaling. To explore whether the NF-κB signaling response reflected antigenic variation, flagellin was purified from 14 different isolates. Purified flagellin filaments generated comparable NF-κB signaling responses, irrespective of either the source of the isolate or H-serotype. These data argued against any variability between isolates being related to flagellin itself. Investigations also argued that neither TLR4 dependent recognition of bacterial lipopolysaccharide nor growth fitness of the isolates played key roles in leading to the variable host response. To determine the roles, if any, of flagellar abundance in inducing these variable responses, flagellar hook numbers of a range of cystitis and ABU isolates were quantified. Images suggested that up to 60% of the isolate population exhibited flagella with the numbers averaging between 1 and 2 flagella per bacterial cell. These data suggest that selective pressures exist in the urinary tract that allow uro-associated E. coli strains to maintain motility, but exploit population heterogeneity, which together function to prevent host TLR5 recognition and bacterial killing.

9.
Mol Microbiol ; 79(1): 264-78, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21166907

ABSTRACT

The flagellar genes in Salmonella enterica are expressed in a temporal hierarchy that mirrors the assembly process itself. The σ(28)-FlgM regulatory circuit plays a key role in controlling this temporal hierarchy. This circuit ensures that the class 3 genes are expressed only when the hook-basal body (HBB), a key intermediate in flagellar assembly, is complete. In this work, we investigated the role of the σ(28)-FlgM regulatory circuit in controlling the timing and magnitude of class 3 gene expression using a combination of mathematical modelling and experimental analysis. Analysis of the model predicted that this circuit continuously controls class 3 gene expression in response to HBB abundance. We experimentally validated these predictions by eliminating different components of the σ(28)-FlgM regulatory system and also by rewiring the transcriptional hierarchy. Based on these results, we conclude that the σ(28)-FlgM regulatory circuit continuously senses the HBB assembly process and regulates class 3 gene expression and possibly flagellar numbers in response.


Subject(s)
Bacterial Proteins/metabolism , Flagellin/biosynthesis , Gene Expression Regulation, Bacterial , Salmonella typhimurium/physiology , Sigma Factor/metabolism , Flagellin/genetics , Gene Expression Profiling , Models, Theoretical , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Time Factors
10.
Eur Urol Open Sci ; 37: 90-98, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35243393

ABSTRACT

BACKGROUND: The AnTIC trial linked continuous low-dose antibiotic prophylaxis treatments to a lower incidence of symptomatic urinary tract infections (UTIs) among individuals performing clean intermittent self-catheterisation (CISC). OBJECTIVE: To explore potential mechanisms underlying the protective effects of low-dose antibiotic prophylaxis treatments, blood and urine samples and uro-associated Escherichia coli isolates from AnTIC participants were analysed. DESIGN SETTING AND PARTICIPANTS: Blood samples (n = 204) were analysed for TLR gene polymorphisms associated with UTI susceptibility and multiple urine samples (n = 558) were analysed for host urogenital responses. E.coli sequence data for 45 temporal isolates recovered from the urine samples of 16 trial participants in the prophylaxis (n = 9) and no-prophylaxis (n = 7) study arms, and characterised by multidrug resistance (MDR), were used to classify individual strains. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: TLR polymorphism data were analysed using Poisson regression. Concentrations of urine host defence markers were analysed using linear mixed-effects models, which accounted for repeated urine samples. RESULTS AND LIMITATIONS: Urine samples from CISC users, irrespective of antibiotic treatment regimens, were associated with robust urothelial innate responses. No links were identified between TLR genotype and CISC user susceptibility to recurrent UTIs. Microbiological study data were limited to the predominant MDR E. coli population; participants prescribed low-dose prophylactic antibiotics were predominantly colonised by a single uro-associated E. coli strain, while participants given acute antibiotic treatments were each colonised by more than one E. coli strain. CONCLUSIONS: Antibiotic treatments did not impact urogenital responses to infection in CISC users. Host genetics in terms of TLR polymorphisms played no role in determining CISC user susceptibility to or protection from recurrent UTIs. Prophylactic antibiotic treatments associated with MDR E. coli were associated with colonisation by stable uro-associated E. coli genotypes. PATIENT SUMMARY: Our findings show that the natural urogenital defences of clean intermittent self-catheterisation (CISC) users were not impacted by antibiotic treatments. For some CISC users, prophylaxis with low-dose antibiotics selected for a stable, predominantly, Esherichia coli rich uromicrobiota.

11.
J Bacteriol ; 193(11): 2695-707, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21441504

ABSTRACT

Bacterial flagella play key roles in surface attachment and host-bacterial interactions as well as driving motility. Here, we have investigated the ability of Caulobacter crescentus to assemble its flagellar filament from six flagellins: FljJ, FljK, FljL, FljM, FljN, and FljO. Flagellin gene deletion combinations exhibited a range of phenotypes from no motility or impaired motility to full motility. Characterization of the mutant collection showed the following: (i) that there is no strict requirement for any one of the six flagellins to assemble a filament; (ii) that there is a correlation between slower swimming speeds and shorter filament lengths in ΔfljK ΔfljM mutants; (iii) that the flagellins FljM to FljO are less stable than FljJ to FljL; and (iv) that the flagellins FljK, FljL, FljM, FljN, and FljO alone are able to assemble a filament.


Subject(s)
Caulobacter crescentus/physiology , Flagella/metabolism , Flagellin/genetics , Flagellin/metabolism , Macromolecular Substances/metabolism , Caulobacter crescentus/genetics , Flagella/ultrastructure , Gene Deletion , Genes, Bacterial , Locomotion , Macromolecular Substances/ultrastructure , Microscopy, Electron
12.
Proteomics ; 11(15): 3036-55, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21726052

ABSTRACT

The endospore-forming Gram-positive pathogen Bacillus anthracis is responsible for the usually fatal disease, inhalational anthrax. The success of this pathogen is dependent on its ability to subvert elements of the innate immune system of its animal hosts. B. anthracis spores, which are the main infective agent, are engulfed and germinate in patrolling alveolar macrophages. In order for the infection to progress, the resulting vegetative cells must resist the antimicrobial oxidative burst mounted by the host NADPH oxidase complex. The response of B. anthracis to this and other macrophage-related stresses is therefore of major importance to the success of this pathogen, and consequently we have analysed the superoxide and peroxide stress stimulons of B. anthracis strain UM23C1-2 by means of a combined transcriptomics and proteomics approach. The results show distinct patterns of expression in response to paraquat (endogenous superoxide) and hydrogen peroxide stress. While the main response to paraquat is the induction of iron uptake pathways, the response to peroxide predominantly involves the induction of protection and repair mechanisms. Comparisons between the responses of B. anthracis and related soil bacterium, B. subtilis, reveal differences that are likely to be relevant to their respective habitats.


Subject(s)
Bacillus anthracis/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Oxidative Stress/physiology , Bacillus anthracis/drug effects , Bacillus anthracis/genetics , Bacillus anthracis/metabolism , Bacillus subtilis/drug effects , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacillus subtilis/physiology , Electrophoresis, Gel, Two-Dimensional , Gene Expression Profiling , Hydrogen Peroxide/pharmacology , Iron/metabolism , Oxidative Stress/drug effects , Paraquat/pharmacology , Proteomics , Siderophores/metabolism
13.
Mol Microbiol ; 78(6): 1416-30, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21143315

ABSTRACT

Each Salmonella enterica serovar Typhimurium cell produces a discrete number of complete flagella. Flagellar assembly responds to changes in growth rates through FlhD(4) C(2) activity. FlhD(4) C(2) activity is negatively regulated by the type 3 secretion chaperone FliT. FliT is known to interact with the flagellar filament cap protein FliD as well as components of the flagellar type 3 secretion apparatus. FliD is proposed to act as an anti-regulator, in a manner similar to FlgM inhibition of σ(28) activity. We have found that efficient growth-dependent regulation of FlhD(4) C(2) requires FliT regulation. In turn, FliD regulation of FliT modulates the response. We also show that, unlike other flagellar-specific regulatory circuits, deletion of fliT or fliD did not lead to an all-or-nothing response in FlhD(4) C(2) activity. To investigate why, we characterized the biochemical interactions in the FliT : FliD : FlhD(4) C(2) circuit. When FlhD(4) C(2) was not bound to DNA, FliT disrupted the FlhD(4) C(2) complex. Interestingly, when FlhD(4) C(2) was bound to DNA it was insensitive to FliT regulation. This suggests that the FliT circuit regulates FlhD(4) C(2) activity by preventing the formation of the FlhD(4) C(2) :DNA complex. Our data would suggest that this level of endogenous regulation of FlhD(4) C(2) activity allows the flagellar system to efficiently respond to external signals.


Subject(s)
Bacterial Proteins/metabolism , Flagella/metabolism , Gene Expression Regulation, Bacterial , Salmonella typhimurium/metabolism , Bacterial Proteins/genetics , Flagella/genetics , Protein Binding , Salmonella typhimurium/genetics , Salmonella typhimurium/growth & development
14.
J Bacteriol ; 192(21): 5767-77, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20833811

ABSTRACT

Salmonella enterica, a common food-borne pathogen, differentially regulates the expression of multiple genes during the infection cycle. These genes encode systems related to motility, adhesion, invasion, and intestinal persistence. Key among them is a type three secretion system (T3SS) encoded within Salmonella pathogenicity island 1 (SPI1). In addition to the SPI1 T3SS, other systems, including flagella and type 1 fimbriae, have been implicated in Salmonella pathogenesis. In this study, we investigated the dynamic expression of the flagellar, SPI1, and type 1 fimbrial genes. We demonstrate that these genes are expressed in a temporal hierarchy, beginning with the flagellar genes, followed by the SPI1 genes, and ending with the type 1 fimbrial genes. This hierarchy could mirror the roles of these three systems during the infection cycle. As multiple studies have shown that extensive regulatory cross talk exists between these three systems, we also tested how removing different regulatory links between them affects gene expression dynamics. These results indicate that cross talk is critical for regulating gene expression during transitional phases in the gene expression hierarchy. In addition, we identified a novel regulatory link between flagellar and type 1 fimbrial gene expression dynamics, where we found that the flagellar regulator, FliZ, represses type 1 fimbrial gene expression through the posttranscriptional regulation of FimZ. The significance of these results is that they provide the first systematic study of the effect of regulatory cross talk on the expression dynamics of flagellar, SPI1, and type 1 fimbrial genes.


Subject(s)
Bacterial Proteins/metabolism , Fimbriae Proteins/metabolism , Gene Expression Regulation, Bacterial/physiology , Salmonella typhimurium/metabolism , Bacterial Proteins/genetics , Fimbriae Proteins/genetics , Mutation , Plasmids , Promoter Regions, Genetic , Salmonella typhimurium/genetics , Signal Transduction , Time Factors
15.
J Bacteriol ; 192(24): 6477-81, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20935096

ABSTRACT

FliZ is an activator of class 2 flagellar gene expression in Salmonella enterica. To understand its role in flagellar assembly, we investigated how FliZ affects gene expression dynamics. We demonstrate that FliZ participates in a positive-feedback loop that induces a kinetic switch in class 2 gene expression.


Subject(s)
Bacterial Proteins/metabolism , Flagella/metabolism , Gene Expression Regulation, Bacterial/physiology , Salmonella enterica/genetics , Salmonella enterica/metabolism , Bacterial Proteins/genetics , Flagella/genetics , Kinetics , Time Factors
16.
Mol Microbiol ; 73(1): 5-19, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19496930

ABSTRACT

This past January, in Cuernavaca Mexico, a conglomerate of scientists met to discuss the contemporary view of Bacterial Locomotion and Signal Transduction (BLAST). The BLAST meetings represent a field that has its roots in chemotaxis and the flagellum-based motility but now encompass all types of cellular movement and signalling. The topics varied from the interactions between molecules to the interactions between species. We heard about 3D reconstructions of transmembrane chemoreceptors within cells, new biophysical methods for understanding cellular engines, intricate phosphorelays, elaborate gene networks, new messenger molecules and emerging behaviours within complex populations of cells. At BLAST X we gained an appreciation for the lifestyle choices bacteria make, how they get to where they are going and the molecular mechanisms that underlie their decisions. Herein we review the highlights of the meeting.


Subject(s)
Bacteria , Bacterial Physiological Phenomena , Locomotion , Signal Transduction , Bacterial Proteins/metabolism , Chemotaxis , Gene Expression Regulation, Bacterial , Membrane Proteins/metabolism , Methyl-Accepting Chemotaxis Proteins
17.
Mol Microbiol ; 70(4): 924-37, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18811728

ABSTRACT

Flagellar gene expression is temporally regulated in response to the assembly state of the growing flagellum. The key mechanism for enforcing this temporal hierarchy in Salmonella enterica serovar Typhimurium is the sigma(28)-FlgM checkpoint, which couples the expression of the late flagellar (P(class3)) genes to the completion of the hook-basal body. This checkpoint is triggered when FlgM is secreted from the cell. In addition to the sigma(28)-FlgM checkpoint, a number of other regulatory mechanisms respond to the secretion of late proteins. In this work, we examined how middle (P(class2)) and late (P(class3)) gene expression is affected by late protein secretion. Dynamic analysis of flagellar gene expression identified a novel mechanism where induction of P(class2) activity is delayed either when late protein secretion is abolished or when late protein secretion is increased. Using a number of different approaches, we were able to show that this mechanism did not involve any known flagellar regulator. Furthermore, the changes in P(class2) activity were not correlated with the associated changes in P(class3) activity, which was found to be proportional to late protein secretion rates. Our data indicate that both P(class2) and P(class3) promoters are continuously regulated in response to assembly and late protein secretion rates. These results suggest that flagellar regulation is more complex than previously thought.


Subject(s)
Bacterial Proteins/metabolism , Flagella/genetics , Gene Expression Regulation, Bacterial , Salmonella typhimurium/genetics , Bacterial Proteins/genetics , Feedback, Physiological , Flagella/metabolism , Genes, Bacterial , Genes, Reporter , Mutation , Phenotype , Promoter Regions, Genetic , Protein Biosynthesis , Salmonella typhimurium/metabolism , Time Factors , Transcription, Genetic
18.
Nat Commun ; 10(1): 5254, 2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31748529

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

19.
Nat Commun ; 10(1): 4379, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31558767

ABSTRACT

Recurrent urinary tract infection (rUTI) is a major medical problem, especially in the elderly and infirm, but the nature of the reservoir of organisms responsible for survival and recolonisation after antibiotic treatment in humans is unclear. Here, we demonstrate the presence of cell-wall deficient (L-form) bacteria in fresh urine from 29 out of 30 older patients with rUTI. In urine, E. coli strains from patient samples readily transition from the walled state to L-form during challenge with a cell wall targeting antibiotic. Following antibiotic withdrawal, they then efficiently transition back to the walled state. E. coli switches between walled and L-form states in a zebrafish larva infection model. The results suggest that L-form switching is a physiologically relevant phenomenon that may contribute to the recurrence of infection in older patients with rUTI, and potentially other infections.


Subject(s)
Cell Wall/metabolism , Escherichia coli Infections/diagnosis , Escherichia coli/metabolism , Urinary Tract Infections/diagnosis , Aged , Animals , Anti-Bacterial Agents/therapeutic use , Escherichia coli/drug effects , Escherichia coli/physiology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Humans , Larva/microbiology , Recurrence , Risk Factors , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology , Zebrafish/microbiology
20.
J Bacteriol ; 190(14): 4979-88, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18469103

ABSTRACT

Flagellar assembly proceeds in a sequential manner, beginning at the base and concluding with the filament. A critical aspect of assembly is that gene expression is coupled to assembly. When cells transition from a nonflagellated to a flagellated state, gene expression is sequential, reflecting the manner in which the flagellum is made. A key mechanism for establishing this temporal hierarchy is the sigma(28)-FlgM checkpoint, which couples the expression of late flagellar (P(class3)) genes to the completion of the hook-basal body. In this work, we investigated the role of FliZ in coupling middle flagellar (P(class2)) gene expression to assembly in Salmonella enterica serovar Typhimurium. We demonstrate that FliZ is an FlhD(4)C(2)-dependent activator of P(class2)/middle gene expression. Our results suggest that FliZ regulates the concentration of FlhD(4)C(2) posttranslationally. We also demonstrate that FliZ functions independently of the flagellum-specific sigma factor sigma(28) and the filament-cap chaperone/FlhD(4)C(2) inhibitor FliT. Furthermore, we show that the previously described ability of sigma(28) to activate P(class2)/middle gene expression is, in fact, due to FliZ, as both are expressed from the same overlapping P(class2) and P(class3) promoters at the fliAZY locus. We conclude by discussing the role of FliZ regulation with respect to flagellar biosynthesis based on our characterization of gene expression and FliZ's role in swimming and swarming motility.


Subject(s)
Flagella/genetics , Gene Expression Regulation, Bacterial , Salmonella typhimurium/physiology , Trans-Activators/metabolism , Artificial Gene Fusion , Bacterial Proteins/metabolism , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Locomotion , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Chaperones/metabolism , Salmonella typhimurium/genetics , Sigma Factor/metabolism , Trans-Activators/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL