Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
PLoS Pathog ; 18(3): e1010423, 2022 03.
Article in English | MEDLINE | ID: mdl-35353877

ABSTRACT

Serratia marcescens is a versatile opportunistic pathogen that can cause a variety of infections, including bacteremia. Our previous work established that the capsule polysaccharide (CPS) biosynthesis and translocation locus contributes to the survival of S. marcescens in a murine model of bacteremia and in human serum. In this study, we determined the degree of capsule genetic diversity among S. marcescens isolates. Capsule loci (KL) were extracted from >300 S. marcescens genome sequences and compared. A phylogenetic comparison of KL sequences demonstrated a substantial level of KL diversity within S. marcescens as a species and a strong delineation between KL sequences originating from infection isolates versus environmental isolates. Strains from five of the identified KL types were selected for further study and electrophoretic analysis of purified CPS indicated the production of distinct glycans. Polysaccharide composition analysis confirmed this observation and identified the constituent monosaccharides for each strain. Two predominant infection-associated clades, designated KL1 and KL2, emerged from the capsule phylogeny. Bacteremia strains from KL1 and KL2 were determined to produce ketodeoxynonulonic acid and N-acetylneuraminic acid, two sialic acids that were not found in strains from other clades. Further investigation of KL1 and KL2 sequences identified two genes, designated neuA and neuB, that were hypothesized to encode sialic acid biosynthesis functions. Disruption of neuB in a KL1 isolate resulted in the loss of sialic acid and CPS production. The absence of sialic acid and CPS production also led to increased susceptibility to internalization by a human monocytic cell line, demonstrating that S. marcescens phagocytosis resistance requires CPS. Together, these results establish the capsule genetic repertoire of S. marcescens and identify infection-associated clades with sialic acid CPS components.


Subject(s)
Bacteremia , Serratia Infections , Animals , Humans , Mice , N-Acetylneuraminic Acid , Phylogeny , Serratia marcescens/genetics
2.
Infect Immun ; 90(2): e0027521, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34871042

ABSTRACT

Uropathogenic Escherichia coli (UPEC) causes the majority of uncomplicated urinary tract infections (UTI), which affect nearly half of women worldwide. Many UPEC strains carry an annotated intimin-like adhesin (ila) locus in their genome related to a well-characterized virulence factor in diarrheagenic E. coli pathotypes. Its role in UPEC uropathogenesis, however, remains unknown. In prototype UPEC strain CFT073, there is an ila locus that contains three predicted intimin-like genes, sinH, sinI, and ratA. We used in silico approaches to determine the phylogeny and genomic distribution of this locus among uropathogens. We found that the currently annotated intimin locus-encoded proteins in CFT073 are more closely related to invasin proteins found in Salmonella. Deletion of the individual sinH, sinI, and ratA genes did not result in measurable effects on growth, biofilm formation, or motility in vitro. On average, sinH was more highly expressed in clinical strains during active human UTI than in human urine ex vivo. Unexpectedly, we found that strains lacking this ila locus had increased adherence to bladder cells in vitro, coupled with a decrease in bladder cell invasion and death. The sinH mutant displayed a significant fitness defect in the murine model of ascending UTI, including reduced inflammation in the bladder. These data confirmed an inhibitory role in bladder cell adherence to facilitate invasion and inflammation; therefore, the ila locus should be termed invasin-like rather than intimin-like. Collectively, our data suggest that loss of this locus mediates measurable interactions with bladder cells in vitro and contributes to fitness during UTI.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Urinary Tract Infections , Uropathogenic Escherichia coli , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Animals , Escherichia coli Infections/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Genomic Islands/genetics , Humans , Inflammation/genetics , Male , Mice , Urinary Tract Infections/genetics , Urothelium
3.
Appl Environ Microbiol ; 86(13)2020 06 17.
Article in English | MEDLINE | ID: mdl-32358013

ABSTRACT

Urinary tract infections (UTI), the second most diagnosed infectious disease worldwide, are caused primarily by uropathogenic Escherichia coli (UPEC), placing a significant financial burden on the health care system. High-throughput transposon mutagenesis combined with genome-targeted sequencing is a powerful technique to interrogate genomes for fitness genes. Genome-wide analysis of E. coli requires random libraries of at least 50,000 mutants to achieve 99.99% saturation; however, the traditional murine model of ascending UTI does not permit testing of large mutant pools due to a bottleneck during infection. To address this, an E. coli CFT073 transposon mutant ordered library of 9,216 mutants was created and insertion sites were identified. A single transposon mutant was selected for each gene to assemble a condensed library consisting of 2,913 unique nonessential mutants. Using a modified UTI model in BALB/c mice, we identified 36 genes important for colonizing the bladder, including purB, yihE, and carB Screening of the condensed library in vitro identified yigP and ubiG to be essential for growth in human urine. Additionally, we developed a novel quantitative PCR (qPCR) technique to identify genes with fitness defects within defined subgroups of related genes (e.g., genes encoding fimbriae, toxins, etc.) following UTI. The number of mutants within these subgroups circumvents bottleneck restriction and facilitates validation of multiple mutants to generate individual competitive indices. Collectively, this study investigates the bottleneck effects during UTI, provides two techniques for evading those effects that can be applied to other disease models, and contributes a genetic tool in prototype strain CFT073 to the field.IMPORTANCE Uropathogenic Escherichia coli strains cause most uncomplicated urinary tract infections (UTI), one of the most common infectious diseases worldwide. Random transposon mutagenesis techniques have been utilized to identify essential bacterial genes during infection; however, this has been met with limitations when applied to the murine UTI model. Conventional high-throughput transposon mutagenesis screens are not feasible because of inoculum size restrictions due to a bottleneck during infection. Our study utilizes a condensed ordered transposon library, limiting the number of mutants while maintaining the largest possible genome coverage. Screening of this library in vivo, and in human urine in vitro, identified numerous candidate fitness factors. Additionally, we have developed a novel technique using qPCR to quantify bacterial outputs following infection with small subgroups of transposon mutants. Molecular approaches developed in this study will serve as useful tools to probe in vivo models that are restricted by anatomical, physiological, or genetic bottleneck limitations.


Subject(s)
DNA Transposable Elements , Escherichia coli Infections/microbiology , Gene Library , Genetic Fitness/physiology , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/genetics , Animals , Female , Humans , Mice , Mice, Inbred BALB C
4.
mBio ; 11(2)2020 04 28.
Article in English | MEDLINE | ID: mdl-32345645

ABSTRACT

Urinary tract infections (UTI) affect half of all women at least once during their lifetime. The rise in the numbers of extended-spectrum beta-lactamase-producing strains and the potential for carbapenem resistance within uropathogenic Escherichia coli (UPEC), the most common causative agent of UTI, create an urgent need for vaccine development. Intranasal immunization of mice with UPEC outer membrane iron receptors FyuA, Hma, IreA, and IutA, conjugated to cholera toxin, provides protection in the bladder or kidneys under conditions of challenge with UPEC strain CFT073 or strain 536. On the basis of these data, we sought to optimize the vaccination route (intramuscular, intranasal, or subcutaneous) in combination with adjuvants suitable for human use, including aluminum hydroxide gel (alum), monophosphoryl lipid A (MPLA), unmethylated CpG synthetic oligodeoxynucleotides (CpG), polyinosinic:polycytidylic acid (polyIC), and mutated heat-labile E. coli enterotoxin (dmLT). Mice intranasally vaccinated with dmLT-IutA and dmLT-Hma displayed significant reductions in bladder colonization (86-fold and 32-fold, respectively), with 40% to 42% of mice having no detectable CFU. Intranasal vaccination of mice with CpG-IutA and polyIC-IutA significantly reduced kidney colonization (131-fold) and urine CFU (22-fold), respectively. dmLT generated the most consistently robust antibody response in intranasally immunized mice, while MPLA and alum produced greater concentrations of antigen-specific serum IgG with intramuscular immunization. On the basis of these results, we conclude that intranasal administration of Hma or IutA formulated with dmLT adjuvant provides the greatest protection from UPEC UTI. This report advances our progress toward a vaccine against uncomplicated UTI, which will significantly improve the quality of life for women burdened by recurrent UTI and enable better antibiotic stewardship.IMPORTANCE Urinary tract infections (UTI) are among the most common bacterial infection in humans, affecting half of all women at least once during their lifetimes. The rise in antibiotic resistance and health care costs emphasizes the need to develop a vaccine against the most common UTI pathogen, Escherichia coli Vaccinating mice intranasally with a detoxified heat-labile enterotoxin and two surface-exposed receptors, Hma or IutA, significantly reduced bacterial burden in the bladder. This work highlights progress in the development of a UTI vaccine formulated with adjuvants suitable for human use and antigens that encode outer membrane iron receptors required for infection in the iron-limited urinary tract.


Subject(s)
Administration, Intranasal , Escherichia coli Proteins/immunology , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli/immunology , Vaccines/therapeutic use , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Bacterial/blood , Bacterial Outer Membrane Proteins/immunology , Drug Administration Routes , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Escherichia coli Infections/therapy , Female , Humans , Immunization/methods , Mice , Receptors, Cell Surface/immunology , Urinary Tract Infections/microbiology , Urinary Tract Infections/therapy , Uropathogenic Escherichia coli/pathogenicity , Vaccination/methods , Vaccines/administration & dosage
5.
PLoS Pathog ; 16(2): e1008382, 2020 02.
Article in English | MEDLINE | ID: mdl-32106241

ABSTRACT

The energy required for a bacterium to grow and colonize the host is generated by metabolic and respiratory functions of the cell. Proton motive force, produced by these processes, drives cellular mechanisms including redox balance, membrane potential, motility, acid resistance, and the import and export of substrates. Previously, disruption of succinate dehydrogenase (sdhB) and fumarate reductase (frdA) within the oxidative and reductive tricarboxylic acid (TCA) pathways in uropathogenic E. coli (UPEC) CFT073 indicated that the oxidative, but not the reductive TCA pathway, is required for fitness in the urinary tract. Those findings led to the hypothesis that fumA and fumC encoding fumarase enzymes of the oxidative TCA cycle would be required for UPEC colonization, while fumB of the reductive TCA pathway would be dispensable. However, only UPEC strains lacking fumC had a fitness defect during experimental urinary tract infection (UTI). To further characterize the role of respiration in UPEC during UTI, additional mutants disrupting both the oxidative and reductive TCA pathways were constructed. We found that knock-out of frdA in the sdhB mutant strain background ameliorated the fitness defect observed in the bladder and kidneys for the sdhB mutant strain and results in a fitness advantage in the bladder during experimental UTI. The fitness defect was restored in the sdhBfrdA double mutant by complementation with frdABCD. Taken together, we demonstrate that it is not the oxidative or reductive pathway that is important for UPEC fitness per se, but rather only the oxidative TCA enzyme FumC. This fumarase lacks an iron-sulfur cluster and is required for UPEC fitness during UTI, most likely acting as a counter measure against exogenous stressors, especially in the iron-limited bladder niche.


Subject(s)
Fumarate Hydratase/metabolism , Iron/metabolism , Uropathogenic Escherichia coli/metabolism , Animals , Citric Acid Cycle/physiology , Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Female , Gene Expression Regulation, Bacterial/physiology , Mice , Mice, Inbred CBA , Oxidation-Reduction , Oxidative Stress , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/physiology
6.
Nat Microbiol ; 5(1): 116-125, 2020 01.
Article in English | MEDLINE | ID: mdl-31686025

ABSTRACT

Metabolic reprogramming is associated with the adaptation of host cells to the disease environment, such as inflammation and cancer. However, little is known about microbial metabolic reprogramming or the role it plays in regulating the fitness of commensal and pathogenic bacteria in the gut. Here, we report that intestinal inflammation reprograms the metabolic pathways of Enterobacteriaceae, such as Escherichia coli LF82, in the gut to adapt to the inflammatory environment. We found that E. coli LF82 shifts its metabolism to catabolize L-serine in the inflamed gut in order to maximize its growth potential. However, L-serine catabolism has a minimal effect on its fitness in the healthy gut. In fact, the absence of genes involved in L-serine utilization reduces the competitive fitness of E. coli LF82 and Citrobacter rodentium only during inflammation. The concentration of luminal L-serine is largely dependent on dietary intake. Accordingly, withholding amino acids from the diet markedly reduces their availability in the gut lumen. Hence, inflammation-induced blooms of E. coli LF82 are significantly blunted when amino acids-particularly L-serine-are removed from the diet. Thus, the ability to catabolize L-serine increases bacterial fitness and provides Enterobacteriaceae with a growth advantage against competitors in the inflamed gut.


Subject(s)
Diet , Enterobacteriaceae/physiology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Serine/metabolism , Animals , Citrobacter rodentium/genetics , Citrobacter rodentium/growth & development , Citrobacter rodentium/metabolism , Citrobacter rodentium/physiology , Colitis/microbiology , Colitis/pathology , Diet/adverse effects , Enterobacteriaceae/genetics , Enterobacteriaceae/growth & development , Enterobacteriaceae/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli/physiology , Gene Expression Regulation, Bacterial , Intestinal Mucosa/metabolism , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Microbial Interactions , Serine/deficiency , Specific Pathogen-Free Organisms
7.
Methods Mol Biol ; 2021: 61-76, 2019.
Article in English | MEDLINE | ID: mdl-31309496

ABSTRACT

Genetic mutation enables the study of the function of specific genes, particularly when a mutant is compared against its isogenic parent. In Proteus mirabilis bacteria, traditional allelic exchange mutation is labor-intensive and has a high failure rate in some strains. Likewise, there is no working protocol for lambda red recombinase-based mutation in P. mirabilis. Here we describe an alternative method of insertional mutagenesis based on retargeting of group II introns. The protocol includes steps to generate single or multiple mutations, with the possibility to delete intervening sequences of DNA.


Subject(s)
Mutagenesis, Insertional/methods , Proteus mirabilis/genetics , Bacterial Proteins/genetics , Bacteriological Techniques , Transformation, Bacterial
8.
Methods Mol Biol ; 2021: 97-108, 2019.
Article in English | MEDLINE | ID: mdl-31309499

ABSTRACT

More than 500 siderophores that bind ferric iron have been characterized and grouped by type based on their chemical structure. The chrome azurol S (CAS) assay is a universal colorimetric method that detects siderophores independent of their structure. In this assay, siderophores scavenge iron from an Fe-CAS-hexadecyltrimethylammonium bromide complex, and subsequent release of the CAS dye results in a color change from blue to orange. Solution-based experiments with CAS result in a quantitative measure of siderophore production, while an observable color change on CAS agar plates can be performed for qualitative detection of siderophores. Cross-feeding assays are another useful method to detect and characterize siderophores produced by bacteria. Under iron-limiting conditions, cross-feeding assays test the ability of an indicator strain to grow when supplied with a specific siderophore (from a test strain) to which it has a cognate receptor required for import into the cell. The cross-feeding assay can be tested with a variety of wild-type strains, siderophore biosynthesis mutants, and siderophore receptor mutants.


Subject(s)
Hydroxybenzoates/chemistry , Proteus mirabilis/metabolism , Siderophores/analysis , Bacteriological Techniques , Calorimetry , Culture Media/chemistry , Siderophores/chemistry
9.
Methods Mol Biol ; 2021: 109-120, 2019.
Article in English | MEDLINE | ID: mdl-31309500

ABSTRACT

A critical first step in bacterial virulence and colonization is adherence to mucosal surfaces, often mediated by fimbriae and other protein adhesins. Here are described three short methods for studying these surface proteins and their behaviors, using protocols developed for the opportunistic pathogen Proteus mirabilis. Unlike the mannose-binding type 1 fimbriae produced by Escherichia coli, most P. mirabilis strains produce mannose-resistant/Proteus-like (MR/P) fimbriae. Both types of fimbrial production and adhesion can be easily demonstrated by a simple and economical hemagglutination assay which uses a model system of erythrocytes. The second and third fimbrial methods presented here show how to shear surface-exposed proteins and use acid treatment to separate interlocked fimbrial subunits into monomers.


Subject(s)
Fimbriae, Bacterial/metabolism , Hemagglutination Tests/methods , Proteus mirabilis/metabolism , Bacterial Adhesion , Bacteriological Techniques , Shear Strength
10.
Methods Mol Biol ; 2021: 201-215, 2019.
Article in English | MEDLINE | ID: mdl-31309507

ABSTRACT

Proteus mirabilis is a major cause of complicated urinary tract infections (UTIs). P. mirabilis' urease activity hydrolyzes urea and raises urine pH levels, which can catalyze bladder and kidney stone formation. This urolithiasis leads to harder-to-treat infections, possible urinary blockage, and subsequent septicemia. Development of a mucosal vaccine against P. mirabilis urinary tract infections is critical to protect against this potentially deadly infection process. Here, we describe the methodology necessary to produce a vaccine candidate conjugated to cholera toxin, administer the vaccine via the intranasal route, and test efficacy in a murine transurethral P. mirabilis infection model.


Subject(s)
Antibodies, Bacterial/metabolism , Immunotoxins/administration & dosage , Proteus Infections/prevention & control , Proteus mirabilis/immunology , Urinary Tract Infections/prevention & control , Administration, Intranasal , Animals , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Disease Models, Animal , Female , Humans , Immunotoxins/immunology , Mice , Mice, Inbred CBA , Vaccination
11.
J Bacteriol ; 201(16)2019 08 15.
Article in English | MEDLINE | ID: mdl-31160397

ABSTRACT

Bacterial metabolism is necessary for adaptation to the host microenvironment. Flexible metabolic pathways allow uropathogenic Escherichia coli (UPEC) to harmlessly reside in the human intestinal tract and cause disease upon extraintestinal colonization. E. coli intestinal colonization requires carbohydrates as a carbon source, while UPEC extraintestinal colonization requires gluconeogenesis and the tricarboxylic acid cycle. UPEC containing disruptions in two irreversible glycolytic steps involving 6-carbon (6-phosphofructokinase; pfkA) and 3-carbon (pyruvate kinase; pykA) substrates have no fitness defect during urinary tract infection (UTI); however, both reactions are catalyzed by isozymes: 6-phosphofructokinases Pfk1 and Pfk2, encoded by pfkA and pfkB, and pyruvate kinases Pyk II and Pyk I, encoded by pykA and pykF UPEC strains lacking one or both phosphofructokinase-encoding genes (pfkB and pfkA pfkB) and strains lacking one or both pyruvate kinase genes (pykF and pykA pykF) were investigated to determine their regulatory roles in carbon flow during glycolysis by examining their fitness during UTI and in vitro growth requirements. Loss of a single phosphofructokinase-encoding gene has no effect on fitness, while the pfkA pfkB double mutant outcompeted the parental strain in the bladder. A defect in bladder and kidney colonization was observed with loss of pykF, while loss of pykA resulted in a fitness advantage. The pykA pykF mutant was indistinguishable from wild-type in vivo, suggesting that the presence of Pyk II rather than the loss of Pyk I itself is responsible for the fitness defect in the pykF mutant. These findings suggest that E. coli suppresses latent enzymes to survive in the host urinary tract.IMPORTANCE Urinary tract infections are the most frequently diagnosed urologic disease, with uropathogenic Escherichia coli (UPEC) infections placing a significant financial burden on the health care system by generating more than two billion dollars in annual costs. This, in combination with steadily increasing antibiotic resistances to present day treatments, necessitates the discovery of new antimicrobial agents to combat these infections. By broadening our scope beyond the study of virulence properties and investigating bacterial physiology and metabolism, we gain a better understanding of how pathogens use nutrients and compete within host microenvironments, enabling us to cultivate new therapeutics to exploit and target pathogen growth requirements in a specific host environment.


Subject(s)
Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Phosphofructokinase-1/metabolism , Pyruvate Kinase/metabolism , Uropathogenic Escherichia coli/enzymology , Adaptation, Physiological , Animals , Escherichia coli Proteins/genetics , Female , Glucose/metabolism , Glycolysis , Humans , Metabolic Networks and Pathways , Mice , Mice, Inbred CBA , Phosphofructokinase-1/genetics , Pyruvate Kinase/genetics , Urinary Tract/microbiology , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/growth & development , Uropathogenic Escherichia coli/physiology
12.
Mol Microbiol ; 109(6): 745-762, 2018 09.
Article in English | MEDLINE | ID: mdl-29884996

ABSTRACT

Acinetobacter baumannii has emerged as a leading nosocomial pathogen, infecting a wide range of anatomic sites including the respiratory tract and the bloodstream. In addition to being multi-drug resistant, little is known about the molecular basis of A. baumannii pathogenesis. To better understand A. baumannii virulence, a combination of a transposon-sequencing (TraDIS) screen and the neutropenic mouse model of bacteremia was used to identify the full set of fitness genes required during bloodstream infection. The lytic transglycosylase MltB was identified as a critical fitness factor. MltB cleaves the MurNAc-GlcNAc bond of peptidoglycan, which leads to cell wall remodeling. Here we show that MltB is part of a complex network connecting resistance to stresses, membrane homeostasis, biogenesis of pili and in vivo fitness. Indeed, inactivation of mltB not only impaired resistance to serum complement, cationic antimicrobial peptides and oxygen species, but also altered the cell envelope integrity, activated the envelope stress response, drastically reduced the number of pili at the cell surface and finally, significantly decreased colonization of both the bloodstream and the respiratory tract.


Subject(s)
Acinetobacter Infections/pathology , Acinetobacter baumannii/genetics , Acinetobacter baumannii/pathogenicity , Cell Membrane/metabolism , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Acinetobacter Infections/microbiology , Acinetobacter baumannii/metabolism , Animals , Antimicrobial Cationic Peptides/pharmacology , Complement System Proteins/immunology , Female , High-Throughput Nucleotide Sequencing , Mice , Mice, Inbred CBA , Muramic Acids/metabolism , Peptidoglycan/metabolism , Stress, Physiological
13.
mBio ; 9(2)2018 03 06.
Article in English | MEDLINE | ID: mdl-29511075

ABSTRACT

Uropathogenic Escherichia coli (UPEC) strains cause most uncomplicated urinary tract infections (UTIs). These strains are a subgroup of extraintestinal pathogenic E. coli (ExPEC) strains that infect extraintestinal sites, including urinary tract, meninges, bloodstream, lungs, and surgical sites. Here, we hypothesize that UPEC isolates adapt to and grow more rapidly within the urinary tract than other E. coli isolates and survive in that niche. To date, there has not been a reliable method available to measure their growth rate in vivo Here we used two methods: segregation of nonreplicating plasmid pGTR902, and peak-to-trough ratio (PTR), a sequencing-based method that enumerates bacterial chromosomal replication forks present during cell division. In the murine model of UTI, UPEC strain growth was robust in vivo, matching or exceeding in vitro growth rates and only slowing after reaching high CFU counts at 24 and 30 h postinoculation (hpi). In contrast, asymptomatic bacteriuria (ABU) strains tended to maintain high growth rates in vivo at 6, 24, and 30 hpi, and population densities did not increase, suggesting that host responses or elimination limited population growth. Fecal strains displayed moderate growth rates at 6 hpi but did not survive to later times. By PTR, E. coli in urine of human patients with UTIs displayed extraordinarily rapid growth during active infection, with a mean doubling time of 22.4 min. Thus, in addition to traditional virulence determinants, including adhesins, toxins, iron acquisition, and motility, very high growth rates in vivo and resistance to the innate immune response appear to be critical phenotypes of UPEC strains.IMPORTANCE Uropathogenic Escherichia coli (UPEC) strains cause most urinary tract infections in otherwise healthy women. While we understand numerous virulence factors are utilized by E. coli to colonize and persist within the urinary tract, these properties are inconsequential unless bacteria can divide rapidly and survive the host immune response. To determine the contribution of growth rate to successful colonization and persistence, we employed two methods: one involving the segregation of a nonreplicating plasmid in bacteria as they divide and the peak-to-trough ratio, a sequencing-based method that enumerates chromosomal replication forks present during cell division. We found that UPEC strains divide extraordinarily rapidly during human UTIs. These techniques will be broadly applicable to measure in vivo growth rates of other bacterial pathogens during host colonization.


Subject(s)
Escherichia coli Infections/genetics , Urinary Tract Infections/genetics , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Humans , Plasmids/genetics
14.
PLoS Pathog ; 13(11): e1006729, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29155899

ABSTRACT

Type VI secretion systems (T6SS) function to deliver lethal payloads into target cells. Many studies have shown that protection against a single, lethal T6SS effector protein requires a cognate antidote immunity protein, both of which are often encoded together in a two-gene operon. The T6SS and an effector-immunity pair is sufficient for both killing and immunity. HereIn this paper we describe a T6SS effector operon that differs from conventional effector-immunity pairs in that eight genes are necessary for lethal effector function, yet can be countered by a single immunity protein. In this study, we investigated the role that the PefE T6SS immunity protein plays in recognition between two strains harboring nearly identical effector operons. Interestingly, despite containing seven of eight identical effector proteins, the less conserved immunity proteins only provided protection against their native effectors, suggesting that specificity and recognition could be dependent on variation within an immunity protein and one effector gene product. The variable effector gene product, PefD, is encoded upstream from pefE, and displays toxic activity that can be countered by PefE independent of T6SS-activity. Interestingly, while the entire pef operon was necessary to exert toxic activity via the T6SS in P. mirabilis, production of PefD and PefE alone was unable to exert this effector activity. Chimeric PefE proteins constructed from two P. mirabilis strains were used to localize immunity function to three amino acids. A promiscuous immunity protein was created using site-directed mutagenesis to change these residues from one variant to another. These findings support the notion that subtle differences between conserved effectors are sufficient for T6SS-mediated kin discrimination and that PefD requires additional factors to function as a T6SS-dependent effector.


Subject(s)
Operon , Type VI Secretion Systems/genetics , Type VI Secretion Systems/immunology , Vibrio cholerae/genetics , Bacterial Proteins/metabolism , Escherichia coli/metabolism , Mutagenesis, Site-Directed/methods , Proteus mirabilis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Type VI Secretion Systems/metabolism , Vibrio cholerae/immunology
15.
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
16.
PLoS Pathog ; 11(1): e1004601, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25568946

ABSTRACT

The human genitourinary tract is a common anatomical niche for polymicrobial infection and a leading site for the development of bacteremia and sepsis. Most uncomplicated, community-acquired urinary tract infections (UTI) are caused by Escherichia coli, while another bacterium, Proteus mirabilis, is more often associated with complicated UTI. Here, we report that uropathogenic E. coli and P. mirabilis have divergent requirements for specific central pathways in vivo despite colonizing and occupying the same host environment. Using mutants of specific central metabolism enzymes, we determined glycolysis mutants lacking pgi, tpiA, pfkA, or pykA all have fitness defects in vivo for P. mirabilis but do not affect colonization of E. coli during UTI. Similarly, the oxidative pentose phosphate pathway is required only for P. mirabilis in vivo. In contrast, gluconeogenesis is required only for E. coli fitness in vivo. The remarkable difference in central pathway utilization between E. coli and P. mirabilis during experimental UTI was also observed for TCA cycle mutants in sdhB, fumC, and frdA. The distinct in vivo requirements between these pathogens suggest E. coli and P. mirabilis are not direct competitors within host urinary tract nutritional niche. In support of this, we found that co-infection with E. coli and P. mirabilis wild-type strains enhanced bacterial colonization and persistence of both pathogens during UTI. Our results reveal that complementary utilization of central carbon metabolism facilitates polymicrobial disease and suggests microbial activity in vivo alters the host urinary tract nutritional niche.


Subject(s)
Coinfection/metabolism , Glycolysis/physiology , Nutritional Physiological Phenomena , Urinary Tract Infections/metabolism , Animals , Coinfection/genetics , Coinfection/microbiology , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/pathogenicity , Escherichia coli Infections/complications , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Female , Glycolysis/genetics , Humans , Mice , Mice, Inbred CBA , Proteus Infections/complications , Proteus Infections/metabolism , Proteus Infections/microbiology , Proteus mirabilis/enzymology , Proteus mirabilis/genetics , Proteus mirabilis/pathogenicity , Transaldolase/genetics , Urinary Tract Infections/microbiology
17.
Infect Immun ; 83(4): 1443-50, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25624354

ABSTRACT

The emergence and spread of extended-spectrum beta-lactamases and carbapenemases among common bacterial pathogens are threatening our ability to treat routine hospital- and community-acquired infections. With the pipeline for new antibiotics virtually empty, there is an urgent need to develop novel therapeutics. Bacteria require iron to establish infection, and specialized pathogen-associated iron acquisition systems like yersiniabactin, common among pathogenic species in the family Enterobacteriaceae, including multidrug-resistant Klebsiella pneumoniae and pathogenic Escherichia coli, represent potentially novel therapeutic targets. Although the yersiniabactin system was recently identified as a vaccine target for uropathogenic E. coli (UPEC)-mediated urinary tract infection (UTI), its contribution to UPEC pathogenesis is unknown. Using an E. coli mutant (strain 536ΔfyuA) unable to acquire yersiniabactin during infection, we established the yersiniabactin receptor as a UPEC virulence factor during cystitis and pyelonephritis, a fitness factor during bacteremia, and a surface-accessible target of the experimental FyuA vaccine. In addition, we determined through transcriptome sequencing (RNA-seq) analyses of RNA from E. coli causing cystitis in women that iron acquisition systems, including the yersiniabactin system, are highly expressed by bacteria during natural uncomplicated UTI. Given that yersiniabactin contributes to the virulence of several pathogenic species in the family Enterobacteriaceae, including UPEC, and is frequently associated with multidrug-resistant strains, it represents a promising novel target to combat antibiotic-resistant infections.


Subject(s)
Cystitis/prevention & control , Escherichia coli Proteins/genetics , Phenols/metabolism , Pyelonephritis/prevention & control , Receptors, Cell Surface/genetics , Thiazoles/metabolism , Uropathogenic Escherichia coli/pathogenicity , Animals , Antibodies, Monoclonal/pharmacology , Bacterial Vaccines/immunology , Cystitis/microbiology , Escherichia coli Infections/immunology , Escherichia coli Proteins/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Phenols/antagonists & inhibitors , Phenols/immunology , Pyelonephritis/microbiology , Receptors, Cell Surface/immunology , Thiazoles/antagonists & inhibitors , Thiazoles/immunology , Urinary Tract Infections/microbiology , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/immunology
18.
Proc Natl Acad Sci U S A ; 111(51): 18327-32, 2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25489107

ABSTRACT

Uropathogenic Escherichia coli (UPEC) is the predominant etiological agent of uncomplicated urinary tract infection (UTI), manifested by inflammation of the urinary bladder, in humans and is a major global public health concern. Molecular pathogenesis of UPEC has been primarily examined using murine models of UTI. Translational research to develop novel therapeutics against this major pathogen, which is becoming increasingly antibiotic resistant, requires a thorough understanding of mechanisms involved in pathogenesis during human UTIs. Total RNA-sequencing (RNA-seq) and comparative transcriptional analysis of UTI samples to the UPEC isolates cultured in human urine and laboratory medium were used to identify novel fitness genes that were specifically expressed during human infection. Evidence for UPEC genes involved in ion transport, including copper efflux, nickel and potassium import systems, as key fitness factors in uropathogenesis were generated using an experimental model of UTI. Translational application of this study was investigated by targeting Cus, a bacterial copper efflux system. Copper supplementation in drinking water reduces E. coli colonization in the urinary bladder of mice. Additionally, our results suggest that anaerobic processes in UPEC are involved in promoting fitness during UTI in humans. In summary, RNA-seq was used to establish the transcriptional signature in UPEC during naturally occurring, community acquired UTI in women and multiple novel fitness genes used by UPEC during human infection were identified. The repertoire of UPEC genes involved in UTI presented here will facilitate further translational studies to develop innovative strategies against UTI caused by UPEC.


Subject(s)
Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Genes, Bacterial , Urinary Tract Infections/microbiology , Escherichia coli/physiology , Humans , Urinary Tract Infections/immunology
19.
J Immunol ; 193(12): 6081-9, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25398327

ABSTRACT

Competition for iron is a critical component of successful bacterial infections, but the underlying in vivo mechanisms are poorly understood. We have previously demonstrated that lipocalin 2 (LCN2) is an innate immunity protein that binds to bacterial siderophores and starves them for iron, thus representing a novel host defense mechanism to infection. In the present study we show that LCN2 is secreted by the urinary tract mucosa and protects against urinary tract infection (UTI). We found that LCN2 was expressed in the bladder, ureters, and kidneys of mice subject to UTI. LCN2 was protective with higher bacterial numbers retrieved from bladders of Lcn2-deficient mice than from wild-type mice infected with the LCN2-sensitive Escherichia coli strain H9049. Uropathogenic E. coli mutants in siderophore receptors for salmochelin, aerobactin, or yersiniabactin displayed reduced fitness in wild-type mice, but not in mice deficient of LCN2, demonstrating that LCN2 imparts a selective pressure on bacterial growth in the bladder. In a human cohort of women with recurrent E. coli UTIs, urine LCN2 levels were associated with UTI episodes and with levels of bacteriuria. The number of siderophore systems was associated with increasing bacteriuria during cystitis. Our data demonstrate that LCN2 is secreted by the urinary tract mucosa in response to uropathogenic E. coli challenge and acts in innate immune defenses as a colonization barrier that pathogens must overcome to establish infection.


Subject(s)
Acute-Phase Proteins/genetics , Bacterial Infections/genetics , Lipocalins/genetics , Proto-Oncogene Proteins/genetics , Urinary Bladder/metabolism , Urinary Bladder/microbiology , Urinary Tract Infections/genetics , Urinary Tract Infections/microbiology , Acute-Phase Proteins/metabolism , Adolescent , Adult , Animals , Bacterial Infections/immunology , Bacterial Infections/metabolism , Bacterial Infections/pathology , Bacterial Load , Cystitis/genetics , Cystitis/immunology , Cystitis/metabolism , Cystitis/microbiology , Disease Models, Animal , Escherichia coli , Female , Gene Expression , Humans , Iron/metabolism , Lipocalin-2 , Lipocalins/metabolism , Mice , Middle Aged , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/pathology , Neutrophil Infiltration , Neutrophils/metabolism , Neutrophils/pathology , Proto-Oncogene Proteins/metabolism , Siderophores/metabolism , Urinary Bladder/pathology , Urinary Tract Infections/immunology , Urinary Tract Infections/pathology , Young Adult
20.
PLoS Pathog ; 9(9): e1003608, 2013.
Article in English | MEDLINE | ID: mdl-24039579

ABSTRACT

The Type VI Secretion System (T6SS) functions in bacteria as a contractile nanomachine that punctures and delivers lethal effectors to a target cell. Virtually nothing is known about the lifestyle or physiology that dictates when bacteria normally produce their T6SS, which prevents a clear understanding of how bacteria benefit from its action in their natural habitat. Proteus mirabilis undergoes a characteristic developmental process to coordinate a multicellular swarming behavior and will discriminate itself from another Proteus isolate during swarming, resulting in a visible boundary termed a Dienes line. Using transposon mutagenesis, we discovered that this recognition phenomenon requires the lethal action of the T6SS. All mutants identified in the genetic screen had insertions within a single 33.5-kb region that encodes a T6SS and cognate Hcp-VrgG-linked effectors. The identified T6SS and primary effector operons were characterized by killing assays, by construction of additional mutants, by complementation, and by examining the activity of the type VI secretion system in real-time using live-cell microscopy on opposing swarms. We show that lethal T6SS-dependent activity occurs when a dominant strain infiltrates deeply beyond the boundary of the two swarms. Using this multicellular model, we found that social recognition in bacteria, underlying killing, and immunity to killing all require cell-cell contact, can be assigned to specific genes, and are dependent on the T6SS. The ability to survive a lethal T6SS attack equates to "recognition". In contrast to the current model of T6SS being an offensive or defensive weapon our findings support a preemptive mechanism by which an entire population indiscriminately uses the T6SS for contact-dependent delivery of effectors during its cooperative mode of growth.


Subject(s)
Bacterial Secretion Systems/physiology , Microbial Interactions/physiology , Proteus mirabilis/physiology , DNA Transposable Elements/genetics , Mutagenesis
SELECTION OF CITATIONS
SEARCH DETAIL
...