Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Gut Microbes ; 16(1): 2333463, 2024.
Article in English | MEDLINE | ID: mdl-38545880

ABSTRACT

The ectopic gut colonization by orally derived pathobionts has been implicated in the pathogenesis of various gastrointestinal diseases, including inflammatory bowel disease (IBD). For example, gut colonization by orally derived Klebsiella spp. has been linked to IBD in mice and humans. However, the mechanisms whereby oral pathobionts colonize extra-oral niches, such as the gut mucosa, remain largely unknown. Here, we performed a high-density transposon (Tn) screening to identify genes required for the adaptation of an oral Klebsiella strain to different mucosal sites - the oral and gut mucosae - at the steady state and during inflammation. We find that K. aerogenes, an oral pathobiont associated with both oral and gut inflammation in mice, harbors a newly identified genomic locus named "locus of colonization in the inflamed gut (LIG)" that encodes genes related to iron acquisition (Sit and Chu) and host adhesion (chaperon usher pili [CUP] system). The LIG locus is highly conserved among K. aerogenes strains, and these genes are also present in several other Klebsiella species. The Tn screening revealed that the LIG locus is required for the adaptation of K. aerogenes in its ectopic niche. In particular, we determined K. aerogenes employs a CUP system (CUP1) present in the LIG locus for colonization in the inflamed gut, but not in the oral mucosa. Thus, oral pathobionts likely exploit distinct adaptation mechanisms in their ectopically colonized intestinal niche compared to their native niche.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Humans , Animals , Mice , Klebsiella/genetics , Inflammatory Bowel Diseases/pathology , Inflammation , Mouth Mucosa
2.
Front Cell Infect Microbiol ; 12: 916247, 2022.
Article in English | MEDLINE | ID: mdl-36204636

ABSTRACT

The genome of Mycobacterium tuberculosis (Mtb) harbors the genetic machinery for assembly of the Fimbrial low-molecular-weight protein (Flp) type IV pilus. Presumably, the Flp pilus is essential for pathogenesis. However, it remains unclear whether the pili genes are transcribed in culture or during infection of host cells. This study aimed to shed light on the expression of the Flp pili-assembly genes (tadZ, tadA, tadB, tadC, flp, tadE, and tadF) in Mtb growing under different growth conditions (exponential phase, stationary phase, and dormancy NRP1 and NRP2 phases induced by hypoxia), during biofilm formation, and in contact with macrophages and alveolar epithelial cells. We found that expression of tad/flp genes was significantly higher in the stationary phase than in exponential or NRP1 or NRP2 phases suggesting that the bacteria do not require type IV pili during dormancy. Elevated gene expression levels were recorded when the bacilli were in contact for 4 h with macrophages or epithelial cells, compared to mycobacteria propagated alone in the cultured medium. An antibody raised against a 12-mer peptide derived from the Flp pilin subunit detected the presence of Flp pili on intra- and extracellular bacteria infecting eukaryotic cells. Altogether, these are compelling data showing that the Flp pili genes are expressed during the interaction of Mtb with host cells and highlight a role for Flp pili in colonization and invasion of the host, subsequently promoting bacterial survival during dormancy.


Subject(s)
Fimbriae Proteins , Mycobacterium tuberculosis , Alveolar Epithelial Cells/metabolism , Bacterial Adhesion , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Macrophages/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Operon
3.
Cell Rep ; 40(3): 111093, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858565

ABSTRACT

Pathobionts employ unique metabolic adaptation mechanisms to maximize their growth in disease conditions. Adherent-invasive Escherichia coli (AIEC), a pathobiont enriched in the gut mucosa of patients with inflammatory bowel disease (IBD), utilizes diet-derived L-serine to adapt to the inflamed gut. Therefore, the restriction of dietary L-serine starves AIEC and limits its fitness advantage. Here, we find that AIEC can overcome this nutrient limitation by switching the nutrient source from the diet to the host cells in the presence of mucolytic bacteria. During diet-derived L-serine restriction, the mucolytic symbiont Akkermansia muciniphila promotes the encroachment of AIEC to the epithelial niche by degrading the mucus layer. In the epithelial niche, AIEC acquires L-serine from the colonic epithelium and thus proliferates. Our work suggests that the indirect metabolic network between pathobionts and commensal symbionts enables pathobionts to overcome nutritional restriction and thrive in the gut.


Subject(s)
Escherichia coli Infections , Bacterial Adhesion , Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Expectorants/metabolism , Humans , Intestinal Mucosa/metabolism , Nutrients , Serine/metabolism
4.
Front Cell Infect Microbiol ; 12: 866416, 2022.
Article in English | MEDLINE | ID: mdl-35651758

ABSTRACT

The human gut acts as the main reservoir of microbes and a relevant source of life-threatening infections, especially in immunocompromised patients. There, the opportunistic fungal pathogen Candida albicans adapts to the host environment and additionally interacts with residing bacteria. We investigated fungal-bacterial interactions by coinfecting enterocytes with the yeast Candida albicans and the Gram-negative bacterium Proteus mirabilis resulting in enhanced host cell damage. This synergistic effect was conserved across different P. mirabilis isolates and occurred also with non-albicans Candida species and C. albicans mutants defective in filamentation or candidalysin production. Using bacterial deletion mutants, we identified the P. mirabilis hemolysin HpmA to be the key effector for host cell destruction. Spatially separated coinfections demonstrated that synergism between Candida and Proteus is induced by contact, but also by soluble factors. Specifically, we identified Candida-mediated glucose consumption and farnesol production as potential triggers for Proteus virulence. In summary, our study demonstrates that coinfection of enterocytes with C. albicans and P. mirabilis can result in increased host cell damage which is mediated by bacterial virulence factors as a result of fungal niche modification via nutrient consumption and production of soluble factors. This supports the notion that certain fungal-bacterial combinations have the potential to result in enhanced virulence in niches such as the gut and might therefore promote translocation and dissemination.


Subject(s)
Candida albicans , Coinfection , Candida , Enterocytes , Humans , Proteus mirabilis/genetics
5.
PLoS Pathog ; 16(10): e1008928, 2020 10.
Article in English | MEDLINE | ID: mdl-33027280

ABSTRACT

Gut dysbiosis associated with intestinal inflammation is characterized by the blooming of particular bacteria such as adherent-invasive E. coli (AIEC). However, the precise mechanisms by which AIEC impact on colitis remain largely unknown. Here we show that antibiotic-induced dysbiosis worsened chemically-induced colitis in IL-22-deficient mice, but not in wild-type mice. The increase in intestinal inflammation was associated with the expansion of E. coli strains with genetic and functional features of AIEC. These E. coli isolates exhibited high ability to out compete related bacteria via colicins and resistance to the host complement system in vitro. Mutation of wzy, the lipopolysaccharide O polymerase gene, rendered AIEC more sensitive to the complement system and more susceptible to engulfment and killing by phagocytes while retaining its ability to outcompete related bacteria in vitro. The wzy AIEC mutant showed impaired fitness to colonize the intestine under colitic conditions, but protected mice from chemically-induced colitis. Importantly, the ability of the wzy mutant to protect from colitis was blocked by depletion of complement C3 which was associated with impaired intestinal eradication of AIEC in colitic mice. These studies link surface lipopolysaccharide O-antigen structure to the regulation of colitic activity in commensal AIEC via interactions with the complement system.


Subject(s)
Complement C3/metabolism , Escherichia coli Infections/drug therapy , Inflammation/microbiology , Lipopolysaccharides/chemistry , Animals , Bacterial Adhesion/drug effects , Bacterial Adhesion/physiology , Crohn Disease/microbiology , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli Infections/microbiology , Intestinal Mucosa/microbiology , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL
6.
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
7.
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
8.
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
9.
Vaccine ; 37(35): 4937-4946, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31320216

ABSTRACT

Urinary tract infection (UTI) is most frequently caused by uropathogenic Escherichia coli (UPEC). Our laboratory has been developing an experimental vaccine targeting four UPEC outer membrane receptors involved in iron acquisition - IreA, FyuA, IutA, and Hma - to elicit protection against UTI. These vaccine targets are all expressed in humans during UTI. In the murine model, high titers of antigen-specific serum IgG or bladder IgA correlate with protection against transurethral challenge with UPEC. Our aim was to measure levels of pre-existing serum antibodies to UTI vaccine antigens in our target population. To accomplish this, we obtained sera from 64 consenting female patients attending a clinic for symptoms of cystitis. As a control, we also collected sera from 20 healthy adult male donors with no history of UTI. Total IgG and antigen-specific IgG titers were measured by ELISA. Of the 64 female patients, 29 had significant bacteriuria (>104 cfu/ml urine) and uropathogenic E. coli (UPEC). Thirty-five patients had non-significant bacteriuria (<104 cfu/ml). Antigen-specific IgG titers did not correlate with the presence or absence of the gene encoding the antigen in the infecting strain (when present), but rather titers were proportional to prevalence of genes encoding antigens among representative collections of UPEC isolates. Surprisingly, we obtained similar results when sera from healthy male patients without history of UTI were tested. Thus, unvaccinated adults have non-protective levels of pre-existing antibodies to UTI vaccine antigens, establishing an important baseline for our target population. This suggests that a UTI vaccine would need to boost pre-existing humoral responses beyond these background levels to protect from infection.


Subject(s)
Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Cystitis/immunology , Escherichia coli Infections/immunology , Escherichia coli Proteins/immunology , Adolescent , Adult , Bacterial Vaccines/chemistry , Bacterial Vaccines/immunology , Bacteriuria/immunology , Bacteriuria/microbiology , Cystitis/microbiology , Female , Humans , Immunoglobulin G/blood , Iron/metabolism , Male , Middle Aged , Uropathogenic Escherichia coli/immunology , Young Adult
10.
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
11.
Cell Host Microbe ; 25(4): 473-474, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30974078

ABSTRACT

Phenylketonuria (PKU) is a rare genetic disorder that causes phenylalanine toxicity in the brain. Two studies, Crook et al. (2019), in this issue of Cell Host & Microbe, and Isabella et al. (2018), employ synthetic biology to develop a live bacterial therapeutic to treat PKU and potentially other metabolic diseases.


Subject(s)
Microbiota , Phenylketonurias , Brain , Escherichia coli , Humans , Phenylalanine
12.
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
13.
Microbiol Spectr ; 4(2)2016 04.
Article in English | MEDLINE | ID: mdl-27227310

ABSTRACT

Bacterial type VI secretion systems (T6SSs) function as contractile nanomachines to puncture target cells and deliver lethal effectors. In the 10 years since the discovery of the T6SS, much has been learned about the structure and function of this versatile protein secretion apparatus. Most of the conserved protein components that comprise the T6SS apparatus itself have been identified and ascribed specific functions. In addition, numerous effector proteins that are translocated by the T6SS have been identified and characterized. These protein effectors usually represent toxic cargoes that are delivered by the attacker cell to a target cell. Researchers in the field are beginning to better understand the lifestyle or physiology that dictates when bacteria normally express their T6SS. In this article, we consider what is known about the structure and regulation of the T6SS, the numerous classes of antibacterial effector T6SS substrates, and how the action of the T6SS relates to a given lifestyle or behavior in certain bacteria.


Subject(s)
Bacteria/metabolism , Type VI Secretion Systems/physiology , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/enzymology , Bacteria/pathogenicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/growth & development , Microbial Interactions/physiology , Type VI Secretion Systems/drug effects
14.
Microbiol Spectr ; 3(3)2015 Jun.
Article in English | MEDLINE | ID: mdl-26185076

ABSTRACT

Among common infections, urinary tract infections (UTI) are the most frequently diagnosed urologic disease. The majority of UTIs are caused by uropathogenic Escherichia coli. The primary niche occupied by E. coli is the lower intestinal tract of mammals, where it resides as a beneficial component of the commensal microbiota. Although it is well-known that E. coli resides in the human intestine as a harmless commensal, specific strains or pathotypes have the potential to cause a wide spectrum of intestinal and diarrheal diseases. In contrast, extraintestinal E. coli pathotypes reside harmlessly in the human intestinal microenvironment but, upon access to sites outside of the intestine, become a major cause of human morbidity and mortality as a consequence of invasive UTI (pyelonephritis, bacteremia, or septicemia). Thus, extraintestinal pathotypes like uropathogenic E. coli (UPEC) possess an enhanced ability to cause infection outside of the intestinal tract and colonize the urinary tract, the bloodstream, or cerebrospinal fluid of human hosts. Due to the requirement for these E. coli to replicate in and colonize both the intestine and extraintestinal environments, we posit that physiology and metabolism of UPEC strains is paramount. Here we discuss that the ability to survive in the urinary tract depends as much on bacterial physiology and metabolism as it does on the well-considered virulence determinants.


Subject(s)
Energy Metabolism/physiology , Escherichia coli Infections/pathology , Urinary Tract Infections/microbiology , Urinary Tract/microbiology , Uropathogenic Escherichia coli/pathogenicity , Amino Acids/metabolism , Carbon/metabolism , Escherichia coli Infections/microbiology , Gene Expression Regulation, Bacterial/genetics , Humans , Intestinal Mucosa/microbiology , Iron/metabolism , Urinary Tract Infections/economics , Uropathogenic Escherichia coli/metabolism , Virulence Factors
15.
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
16.
Pathogens ; 5(1)2015 Dec 31.
Article in English | MEDLINE | ID: mdl-26729174

ABSTRACT

Urinary tract infection (UTI) is the second most common infection in humans after those involving the respiratory tract. This results not only in huge annual economic costs, but in decreased workforce productivity and high patient morbidity. Most infections are caused by uropathogenic Escherichia coli (UPEC). Antibiotic treatment is generally effective for eradication of the infecting strain; however, documentation of increasing antibiotic resistance, allergic reaction to certain pharmaceuticals, alteration of normal gut flora, and failure to prevent recurrent infections represent significant barriers to treatment. As a result, approaches to prevent UTI such as vaccination represent a gap that must be addressed. Our laboratory has made progress toward development of a preventive vaccine against UPEC. The long-term research goal is to prevent UTIs in women with recurrent UTIs. Our objective has been to identify the optimal combination of protective antigens for inclusion in an effective UTI vaccine, optimal adjuvant, optimal dose, and optimal route of delivery. We hypothesized that a multi-subunit vaccine elicits antibody that protects against experimental challenge with UPEC strains. We have systematically identified four antigens that can individually protect experimentally infected mice from colonization of the bladder and/or kidneys by UPEC when administered intranasally with cholera toxin (CT) as an adjuvant. To advance the vaccine for utility in humans, we will group the individual antigens, all associated with iron acquisition (IreA, Hma, IutA, FyuA), into an effective combination to establish a multi-subunit vaccine. We demonstrated for all four vaccine antigens that antigen-specific serum IgG represents a strong correlate of protection in vaccinated mice. High antibody titers correlate with low colony forming units (CFUs) of UPEC following transurethral challenge of vaccinated mice. However, the contribution of cell-mediated immunity cannot be ruled out and must be investigated experimentally. We have demonstrated that antibodies bind to the surface of UPEC expressing the antigens. Sera from women with and without histories of UTI have been tested for antibody levels to vaccine antigens. Our results validate iron acquisition as a target for vaccination against UTI.

17.
Microbiologyopen ; 3(5): 630-41, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25100003

ABSTRACT

Swarming contributes to Proteus mirabilis pathogenicity by facilitating access to the catheterized urinary tract. We previously demonstrated that 0.1-20 mmol/L arginine promotes swarming on normally nonpermissive media and that putrescine biosynthesis is required for arginine-induced swarming. We also previously determined that arginine-induced swarming is pH dependent, indicating that the external proton concentration is critical for arginine-dependent effects on swarming. In this study, we utilized survival at pH 5 and motility as surrogates for measuring changes in the proton gradient (ΔpH) and proton motive force (µH(+) ) in response to arginine. We determined that arginine primarily contributes to ΔpH (and therefore µH(+) ) through the action of arginine decarboxylase (speA), independent of the role of this enzyme in putrescine biosynthesis. In addition to being required for motility, speA also contributed to fitness during infection. In conclusion, consumption of intracellular protons via arginine decarboxylase is one mechanism used by P. mirabilis to conserve ΔpH and µH(+) for motility.


Subject(s)
Arginine/metabolism , Proteus mirabilis/cytology , Proteus mirabilis/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Proteus mirabilis/chemistry , Proteus mirabilis/enzymology , Proton-Motive Force , Protons
18.
Infect Immun ; 82(10): 4241-52, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25069986

ABSTRACT

The metV genomic island in the chromosome of uropathogenic Escherichia coli (UPEC) encodes a putative transcription factor and a sugar permease of the phosphotransferase system (PTS), which are predicted to compose a Bgl-like sensory system. The presence of these two genes, hereby termed pafR and pafP, respectively, has been previously shown to correlate with isolates causing clinical syndromes. We show here that deletion of both genes impairs the ability of the resulting mutant to infect the CBA/J mouse model of ascending urinary tract infection compared to that of the parent strain, CFT073. Expressing the two genes in trans in the two-gene knockout mutant complemented full virulence. Deletion of either gene individually generated the same phenotype as the double knockout, indicating that both pafR and pafP are important to pathogenesis. We screened numerous environmental conditions but failed to detect expression from the promoter that precedes the paf genes in vitro, suggesting that they are in vivo induced (ivi). Although PafR is shown here to be capable of functioning as a transcriptional antiterminator, its targets in the UPEC genome are not known. Using microarray analysis, we have shown that expression of PafR from a heterologous promoter in CFT073 affects expression of genes related to bacterial virulence, biofilm formation, and metabolism. Expression of PafR also inhibits biofilm formation and motility. Taken together, our results suggest that the paf genes are implicated in pathogenesis and that PafR controls virulence genes, in particular biofilm formation genes.


Subject(s)
Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Monosaccharide Transport Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Uropathogenic Escherichia coli/pathogenicity , Virulence Factors/metabolism , Animals , Biofilms/growth & development , Disease Models, Animal , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Escherichia coli Proteins/genetics , Female , Gene Deletion , Gene Expression Profiling , Genetic Complementation Test , Locomotion , Mice , Mice, Inbred CBA , Microarray Analysis , Monosaccharide Transport Proteins/genetics , Transcription Factors/genetics , Urinary Tract Infections/microbiology , Urinary Tract Infections/pathology , Uropathogenic Escherichia coli/genetics , Virulence , Virulence Factors/genetics
19.
Infect Immun ; 82(9): 3644-56, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24935980

ABSTRACT

A heterogeneous subset of extraintestinal pathogenic Escherichia coli (ExPEC) strains, referred to as uropathogenic E. coli (UPEC), causes most uncomplicated urinary tract infections. However, no core set of virulence factors exists among UPEC strains. Instead, the focus of the analysis of urovirulence has shifted to studying broad classes of virulence factors and the interactions between them. For example, the RTX nonfimbrial adhesin TosA mediates adherence to host cells derived from the upper urinary tract. The associated tos operon is well expressed in vivo but poorly expressed in vitro and encodes TosCBD, a predicted type 1 secretion system. TosR and TosEF are PapB and LuxR family transcription factors, respectively; however, no role has been assigned to these potential regulators. Thus, the focus of this study was to determine how TosR and TosEF regulate tosA and affect the reciprocal expression of adhesins and flagella. Among a collection of sequenced UPEC strains, 32% (101/317) were found to encode TosA, and nearly all strains (91% [92/101]) simultaneously carried the putative regulatory genes. Deletion of tosR alleviates tosA repression. The tos promoter was localized upstream of tosR using transcriptional fusions of putative promoter regions with lacZ. TosR binds to this region, affecting a gel shift. A 100-bp fragment 220 to 319 bp upstream of tosR inhibits binding, suggesting localization of the TosR binding site. TosEF, on the other hand, downmodulate motility when overexpressed by preventing the expression of fliC, encoding flagellin. Deletion of tosEF increased motility. Thus, we present an additional example of the reciprocal control of adherence and motility.


Subject(s)
Bacterial Toxins/genetics , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial/genetics , Membrane Proteins/genetics , Repressor Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Uropathogenic Escherichia coli/genetics , Adhesins, Bacterial/genetics , Adhesins, Escherichia coli/genetics , Amino Acid Sequence , Binding Sites/genetics , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Flagella/genetics , Flagella/microbiology , Flagellin/genetics , Molecular Sequence Data , Operon/genetics , Promoter Regions, Genetic/genetics , Sequence Alignment , Urinary Tract Infections/genetics , Urinary Tract Infections/microbiology , Virulence Factors/genetics
20.
PLoS Pathog ; 10(5): e1004124, 2014 May.
Article in English | MEDLINE | ID: mdl-24809621

ABSTRACT

SslE, the Secreted and surface-associated lipoprotein from Escherichia coli, has recently been associated to the M60-like extracellular zinc-metalloprotease sub-family which is implicated in glycan recognition and processing. SslE can be divided into two main variants and we recently proposed it as a potential vaccine candidate. By applying a number of in vitro bioassays and comparing wild type, knockout mutant and complemented strains, we have now demonstrated that SslE specifically contributes to degradation of mucin substrates, typically present in the intestine and bladder. Mutation of the zinc metallopeptidase motif of SslE dramatically impaired E. coli mucinase activity, confirming the specificity of the phenotype observed. Moreover, antibodies raised against variant I SslE, cloned from strain IHE3034 (SslEIHE3034), are able to inhibit translocation of E. coli strains expressing different variants through a mucin-based matrix, suggesting that SslE induces cross-reactive functional antibodies that affect the metallopeptidase activity. To test this hypothesis, we used well-established animal models and demonstrated that immunization with SslEIHE3034 significantly reduced gut, kidney and spleen colonization by strains producing variant II SslE and belonging to different pathotypes. Taken together, these data strongly support the importance of SslE in E. coli colonization of mucosal surfaces and reinforce the use of this antigen as a component of a broadly protective vaccine against pathogenic E. coli species.


Subject(s)
Antibodies, Bacterial/pharmacology , Antibody Formation , Escherichia coli Infections , Escherichia coli Proteins/immunology , Polysaccharide-Lyases/antagonists & inhibitors , Virulence Factors/immunology , Animals , Animals, Outbred Strains , Antibodies, Bacterial/metabolism , Cells, Cultured , Enteropathogenic Escherichia coli/growth & development , Enteropathogenic Escherichia coli/immunology , Enteropathogenic Escherichia coli/metabolism , Enzyme Activation/drug effects , Escherichia coli/growth & development , Escherichia coli/immunology , Escherichia coli/metabolism , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Female , Intestines/microbiology , Mice , Mice, Inbred CBA , Polysaccharide-Lyases/immunology , Polysaccharide-Lyases/metabolism , Virulence Factors/antagonists & inhibitors , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...