Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters











Publication year range
1.
PLoS One ; 15(8): e0237135, 2020.
Article in English | MEDLINE | ID: mdl-32822422

ABSTRACT

DNA-binding Transcription Factors (TFs) play a central role in regulation of gene expression in prokaryotic organisms, and similarities at the sequence level have been reported. These proteins are predicted with different abundances as a consequence of genome size, where small organisms contain a low proportion of TFs and large genomes contain a high proportion of TFs. In this work, we analyzed a collection of 668 experimentally validated TFs across 30 different species from diverse taxonomical classes, including Escherichia coli K-12, Bacillus subtilis 168, Corynebacterium glutamicum, and Streptomyces coelicolor, among others. This collection of TFs, together with 111 hidden Markov model profiles associated with DNA-binding TFs collected from diverse databases such as PFAM and DBD, was used to identify the repertoire of proteins putatively devoted to gene regulation in 1321 representative genomes of Archaea and Bacteria. The predicted regulatory proteins were posteriorly analyzed in terms of their genomic context, allowing the prediction of functions for TFs and their neighbor genes, such as genes involved in virulence, enzymatic functions, phosphorylation mechanisms, and antibiotic resistance. The functional analysis associated with PFAM groups showed diverse functional categories were significantly enriched in the collection of TFs and the proteins encoded by the neighbor genes, in particular, small-molecule binding and amino acid transmembrane transporter activities associated with the LysR family and proteins devoted to cellular aromatic compound metabolic processes or responses to drugs, stress, or abiotic stimuli in the MarR family. We consider that with the increasing data derived from new technologies, novel TFs can be identified and help improve the predictions for this class of proteins in complete genomes. The complete collection of experimentally characterized and predicted TFs is available at http://web.pcyt.unam.mx/EntrafDB/.


Subject(s)
Archaea/genetics , Archaeal Proteins/genetics , Bacterial Proteins/genetics , DNA-Binding Proteins/genetics , Escherichia coli K12/genetics , Transcription Factors/genetics , Archaea/pathogenicity , Archaeal Proteins/metabolism , Bacterial Proteins/metabolism , Binding Sites , DNA, Archaeal/metabolism , DNA, Bacterial/metabolism , DNA-Binding Proteins/metabolism , Escherichia coli K12/pathogenicity , Gene Expression Regulation, Archaeal , Gene Expression Regulation, Bacterial , Genome, Archaeal , Genome, Bacterial , Protein Binding , Transcription Factors/metabolism , Virulence/genetics
2.
Biosens Bioelectron ; 145: 111624, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31546201

ABSTRACT

In spite of the clinical need, there is a major gap in rapid diagnostics for identification and quantitation of E. coli and other pathogens, also regarded as the biggest bottleneck in the fight against the spread of antimicrobial resistant bacterial strains. This study reports for the first time an optical, smartphone-based microfluidic fluorescence sandwich immunoassay capable of quantifying E. coli in buffer and synthetic urine in less than 25 min without sample preparation nor concentration. A limit of detection (LoD) up to 240 CFU/mL, comensurate with cut-off for UTIs (103-105 CFUs/mL) was achieved. Replicas of full response curves performed with 100-107 CFUs/mL of E. coli K12 in synthetic urine yielded recovery values in the range 80-120%, assay reproducibility below 30% and precision below 20%, therefore similar to high-performance automated immunoassays. The unrivalled LoD was mainly linked to the 'open fluidics' nature of the 10-bore microfluidic strips used that enabled passing a large volume of sample through the microcapillaries coated with capture antibody. The new smartphone based test has the potential of being as a rapid, point-of-care test for rule-in of E. coli infections that are responsible for around 80% of UTIs, helping to stop the over-prescription of antibiotics and the monitoring of patients with other symptomatic communicable diseases caused by E. coli at global scale.


Subject(s)
Biosensing Techniques , Escherichia coli K12/isolation & purification , Smartphone , Urinary Tract Infections/microbiology , Escherichia coli K12/pathogenicity , Humans , Immunoassay , Microfluidics
3.
Microb Pathog ; 133: 103545, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31112772

ABSTRACT

Commensal enteric microbes under specific conditions viz. immunocompromised system, altered microbiota or uncompetitive niche induce their otherwise dormant pathogenic phenotype to distort host cellular functioning. Here we investigate how under in vitro environment established by using Caco-2 cells, commensal gut microbe E. coli K12 (ATCC 14849) disrupt intestinal epithelial barrier function. Caco-2 cells exposed to E. coli showed the time dependent significant (P < 0.01) decrease in transepithelial electrical resistance (TEER) and concomitantly increased phenol red flux across cell monolayer in contrast to non infected control cells. E. coli infected intestinal cells were observed with suppressed (p < 0.05) mRNA levels of ZO-1, Claudin-1, Occludin and Cingulin-1 in contrast to significantly (p < 0.05) higher PIgR and hbd-2 mRNA fold changes. Immunofluorescent and electron micrographs revealed the disrupted distribution and localisation of specific tight junction proteins (Zo-1 and Claudin-1) and actin filament in E. coli infected Caco-2 cells that ultimately resulted in deformed cellular morphology. Taken together, E. coli K12 under compromised in vitro milieu disrupted the intestinal barrier functions by decreasing the expression of important tight junction genes along with the altered distribution of associated proteins that increased the intestinal permeability as reflected by phenol red flux and TEER values.


Subject(s)
Escherichia coli K12/physiology , Escherichia coli K12/pathogenicity , Gastrointestinal Microbiome , Opportunistic Infections/microbiology , Symbiosis , Caco-2 Cells/cytology , Caco-2 Cells/microbiology , Claudin-1/metabolism , Cytoskeletal Proteins , Electric Impedance , Epithelial Cells/metabolism , Gene Expression , Host Microbial Interactions , Humans , Intestinal Mucosa/metabolism , Intestines/microbiology , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Occludin/genetics , Occludin/metabolism , Permeability , RNA, Messenger , Tight Junction Proteins/metabolism , Tight Junctions/metabolism , Zonula Occludens-1 Protein/metabolism , beta-Defensins/metabolism
4.
Sci Total Environ ; 657: 918-925, 2019 Mar 20.
Article in English | MEDLINE | ID: mdl-30677957

ABSTRACT

Antibiotic resistant pathogenic bacteria (ARPB) residual in soil-plant system has caused serious threat against public health and environmental safety. Being capable of targeted lysing host bacteria, phage therapy has been proposed as promising method to control the ARPB contamination in environments. In this study, microcosm trials were performed to investigate the impact of various phage treatments on the dissipation of tetracycline resistant Escherichia coli K-12 and chloramphenicol resistant Pseudomonas aeruginosa PAO1 in soil-carrot system. After 70 days of incubation, all the four phage treatments significantly decreased the abundance of the pathogenic bacteria and the corresponding antibiotic resistance genes (tetW and cmlA) in the soil-carrot system (p < 0.05), following the order of the cocktail phage treatment (phages ΦYSZ1 + ΦYSZ2) > the polyvalent phage (ΦYSZ3 phage with broad host range) treatment > host-specific phage (ΦYSZ2 and ΦYSZ1) treatments > the control. In addition, the polyvalent phage treatment also exerted positive impact on the diversity and stability of the bacterial community in the system, suggesting that this is an environmentally friendly technique with broad applications in the biocontrol of ARPB/ARGs in soil-plant system.


Subject(s)
Bacteriophages/physiology , Drug Resistance, Microbial , Escherichia coli K12/virology , Pseudomonas aeruginosa/virology , Soil Microbiology , Biodiversity , Biological Control Agents , Daucus carota/microbiology , Escherichia coli K12/drug effects , Escherichia coli K12/pathogenicity , Microbial Consortia , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/pathogenicity , Tetracycline Resistance
5.
Sci Rep ; 8(1): 16145, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30385858

ABSTRACT

Blood platelets can interact with bacteria, possibly leading to platelet activation, cytokine and microparticle release and immune signalling. Besides, bacteria can also affect the platelet RNA content. We investigated the impact of non-pathogenic K12 and pathogenic O18:K1 Escherichia (E.) coli strains on platelet activation, RNA expression patterns, and selected proteins. Depending on bacteria concentration, contact of platelets with E. coli K12 lead to an increase of P-selectin (24-51.3%), CD63 (15.9-24.3%), PAC-1 (3.8-14.9%) and bound fibrinogen (22.4-39%) on the surface. E. coli O18:K1 did not affect these markers. Sequencing analysis of total RNA showed that E. coli K12 caused a significant concentration change of 103 spliced mRNAs, of which 74 decreased. For the RNAs of HMBS (logFC = +5.73), ATP2C1 (logFC = -3.13) and LRCH4 (logFC = -4.07) changes were detectable by thromboSeq and Tuxedo pipelines. By Western blot we observed the conversion of HMBS protein from a 47 kDA to 40 kDa product by E. coli K12, O18:K1 and by purified lipopolysaccharide. While ATP2C1 protein was released from platelets, E. coli either reduced the secretion or broke down the released protein making it undetectable by antibodies. Our results demonstrate that different E. coli strains influence activation, RNA and protein levels differently which may affect platelet-bacteria crosstalk.


Subject(s)
Blood Platelets/metabolism , Calcium-Transporting ATPases/genetics , Escherichia coli K12/genetics , Nerve Tissue Proteins/genetics , Uroporphyrinogen III Synthetase/genetics , Antigens, Bacterial/genetics , Calcium-Transporting ATPases/blood , Escherichia coli Infections/blood , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Escherichia coli K12/pathogenicity , Gene Expression Regulation, Bacterial/genetics , Humans , Lipopolysaccharides/genetics , P-Selectin/genetics , Platelet Activation/genetics , RNA/blood , RNA/genetics , Sequence Analysis, RNA , Tetraspanin 30/genetics
6.
Infect Immun ; 85(12)2017 12.
Article in English | MEDLINE | ID: mdl-28893912

ABSTRACT

Locus of enterocyte effacement (LEE)-negative Shiga toxin (Stx)-producing Escherichia coli (STEC) strains are human pathogens that lack the LEE locus, a pathogenicity island (PAI) involved in the intimate adhesion of LEE-positive strains to the host gut epithelium. The mechanism used by LEE-negative STEC strains to colonize the host intestinal mucosa is still not clear. The cell invasion determinant tia, previously described in enterotoxigenic E. coli strains, has been identified in LEE-negative STEC strains that possess the subtilase-encoding pathogenicity island (SE-PAI). We evaluated the role of the gene tia, present in these LEE-negative STEC strains, in the invasion of monolayers of cultured cells. We observed that these strains were able to invade Caco-2 and HEp-2 cell monolayers and compared their invasion ability with that of a mutant strain in which the gene tia had been inactivated. Mutation of the gene tia resulted in a strong reduction of the invasive phenotype, and complementation of the tia mutation with a functional copy of the gene restored the invasion activity. Moreover, we show that the gene tia is overexpressed in bacteria actively invading cell monolayers, demonstrating that tia is involved in the ability to invade cultured monolayers of epithelial cells shown by SE-PAI-positive E. coli, including STEC, strains. However, the expression of the tia gene in the E. coli K-12 strain JM109 was not sufficient, in its own right, to confer to this strain the ability to invade cell monolayers, suggesting that at least another factor must be involved in the invasion ability displayed by the SE-PAI-positive strains.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Endocytosis , Epithelial Cells/microbiology , Escherichia coli Proteins/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity , Virulence Factors/metabolism , Cell Line , Escherichia coli K12/genetics , Escherichia coli K12/pathogenicity , Gene Knockout Techniques , Genetic Complementation Test , Genomic Islands , Humans , Shiga-Toxigenic Escherichia coli/genetics
7.
Appl Microbiol Biotechnol ; 100(21): 8985-9001, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27645299

ABSTRACT

Acetate is ubiquitously found in natural environments. Its availability in the gut is high as a result of the fermentation of nutrients, and although it is rapidly absorbed by intestinal mucosa, it can also be used as carbon source by some members of gut microbiota. The metabolism of acetate in Escherichia coli has attracted the attention of the scientific community due to its role in central metabolism and its link to multiple physiological features. In this microorganism, acetate is involved directly or indirectly on the regulation of functional processes, such as motility, formation of biofilms, and responses to stress. Furthermore, it is a relevant nutrient in gut, where it serves additional roles, which regulate or, at least, modulate pathophysiological responses of E. coli and other bacteria. Acetate is one of the major by-products of anaerobic (fermenting) metabolism, and it is also produced under fully aerobic conditions. This acetate overflow is recognized as one of the major drawbacks limiting E. coli's productivity in biotechnological processes. This review sums up current knowledge on acetate metabolism in E. coli, explaining the major milestones that have led to deciphering its complex regulation in the K-12 strain. Major differences in the metabolism of acetate in other strains will be underlined, with a focus on strains of biotechnological and biomedical interest.


Subject(s)
Acetates/metabolism , Carbon/metabolism , Escherichia coli K12/genetics , Escherichia coli K12/metabolism , Gene Expression Regulation, Bacterial , Escherichia coli K12/pathogenicity
8.
Sci Rep ; 6: 31722, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27530146

ABSTRACT

To explore the interaction of gut microbes and the host immune system, bacteria were isolated from the gut of Protaetia brevitarsis seulensis larvae. Chryseobacterium sp., Bacillus subtilis, Arthrobacter arilaitensis, Bacillus amyloliquefaciens, Bacillus megaterium, and Lysinibacillus xylanilyticus were cultured in vitro, identified, and injected in the hemocoel of P. brevitarsis seulensis larvae, respectively. There were no significant changes in phagocytosis-associated lysosomal formation or pathogen-related autophagosome in immune cells (granulocytes) from Chryseobacterium sp.-challenged larvae. Next, we examined changes in the transcription of innate immune genes such as peptidoglycan recognition proteins and antimicrobial peptides following infection with Chryseobacterium sp. PGRP-1 and -2 transcripts, which may be associated with melanization generated by prophenoloxidase (PPO), were either highly or moderately expressed at 24 h post-infection with Chryseobacterium sp. However, PGRP-SC2 transcripts, which code for bactericidal amidases, were expressed at low levels. With respect to antimicrobial peptides, only coleoptericin was moderately expressed in Chryseobacterium sp.-challenged larvae, suggesting maintenance of an optimum number of Chryseobacterium sp. All examined genes were expressed at significantly higher levels in larvae challenged with a pathogenic bacterium. Our data demonstrated that gut-inhabiting bacteria, the Chryseobacterium sp., induced a weaker immune response than other pathogenic bacteria, E. coli K12.


Subject(s)
Chryseobacterium/immunology , Chryseobacterium/pathogenicity , Coleoptera/immunology , Coleoptera/microbiology , Animals , Autophagosomes/immunology , Autophagosomes/microbiology , Chryseobacterium/classification , Coleoptera/genetics , Escherichia coli K12/immunology , Escherichia coli K12/pathogenicity , Gastrointestinal Microbiome/immunology , Gene Expression , Genes, Insect , Granulocytes/immunology , Granulocytes/microbiology , Hemocytes/immunology , Hemocytes/microbiology , Host-Pathogen Interactions/immunology , Immune Tolerance/genetics , Immunity, Cellular/genetics , Larva/genetics , Larva/immunology , Larva/microbiology , Lysosomes/immunology , Lysosomes/microbiology , Phylogeny
9.
Biochem J ; 466(2): 323-35, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25484033

ABSTRACT

Pet is a cytotoxic autotransporter protein secreted by the pathogenic enteroaggregative Escherichia coli strain 042. Expression of Pet is co-dependent on two global transcription regulators: CRP (cyclic AMP receptor protein) and Fis (factor for inversion stimulation). At the pet promoter CRP binds to a single site centred at position -40.5 upstream of the start site for transcription. Due to the suboptimal positioning of this site, CRP alone activates transcription poorly and requires Fis to bind upstream to promote full activation. Here, we show that CRP and Fis control the expression of other important autotransporter toxins, namely Sat from uropathogenic E. coli (UPEC) and SigA from Shigella sonnei, and that this regulation has been conserved in different pathogens. Furthermore, we investigate the mechanism of Fis-mediated co-activation, exploiting a series of semi-synthetic promoters, with similar architecture to the pet promoter. We show that, when bound at position -40.5, CRP recruits RNA polymerase inefficiently and that Fis compensates by aiding polymerase recruitment through a direct protein-protein interaction. We demonstrate that other suitably positioned upstream transcription factors, which directly recruit RNA polymerase, can also compensate for the inappropriate positioning of CRP. We propose that this is a simple 'shared-recruitment' mechanism, by which co-dependence of promoters on two transcription factors could evolve.


Subject(s)
Bacterial Toxins/metabolism , Cyclic AMP Receptor Protein/metabolism , Escherichia coli Proteins/metabolism , Factor For Inversion Stimulation Protein/metabolism , Gene Expression Regulation, Bacterial , Models, Molecular , Response Elements , Uropathogenic Escherichia coli/metabolism , 5' Flanking Region , Amino Acid Substitution , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Cyclic AMP Receptor Protein/chemistry , Cyclic AMP Receptor Protein/genetics , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Electrophoretic Mobility Shift Assay , Enterotoxins/genetics , Enterotoxins/metabolism , Escherichia coli K12/enzymology , Escherichia coli K12/metabolism , Escherichia coli K12/pathogenicity , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Factor For Inversion Stimulation Protein/chemistry , Factor For Inversion Stimulation Protein/genetics , Mutation , Promoter Regions, Genetic , Protein Interaction Domains and Motifs , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Shigella sonnei/enzymology , Shigella sonnei/metabolism , Shigella sonnei/pathogenicity , Sigma Factor/chemistry , Sigma Factor/genetics , Sigma Factor/metabolism , Transcription, Genetic , Uropathogenic Escherichia coli/enzymology , Uropathogenic Escherichia coli/pathogenicity
10.
Cell Host Microbe ; 15(6): 779-91, 2014 Jun 11.
Article in English | MEDLINE | ID: mdl-24882704

ABSTRACT

Adult hematopoietic stem cells (HSCs) are maintained in specialized niches within the bone marrow under steady-state conditions and mobilize for extramedullary hematopoiesis during periods of stress such as bacterial infections. However, the underlying mechanisms are unclear. We show that systemic infection of mice with Escherichia coli, commonly associated with bacteremia in humans, mobilizes functional HSCs to the spleen. Accumulation of splenic HSCs (CD150+CD48-Lin(-/low)Sca1+cKit+) was diminished in TLR4-deficient and RIPK2-deficient mice, implicating TLRs and cytosolic NOD1/NOD2 signaling in the process. Accordingly, dual stimulation of NOD1 and TLR4 in radio-resistant cells alone was sufficient to mobilize HSCs, while TLR4 expression on HSCs was dispensable. Mechanistically, TLR4 and NOD1 synergistically induced granulocyte colony-stimulating factor (G-CSF), which was required for extramedullary HSC accumulation. Mobilized HSCs and progenitor cells gave rise to neutrophils and monocytes and contributed to limiting secondary infection.


Subject(s)
Escherichia coli Infections/metabolism , Hematopoietic Stem Cells/microbiology , Nod1 Signaling Adaptor Protein/metabolism , Toll-Like Receptor 4/metabolism , Animals , Antigens, CD/metabolism , Ataxin-10 , Bone Marrow/microbiology , Bone Marrow/pathology , CD48 Antigen , Carrier Proteins/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Escherichia coli K12/pathogenicity , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Spleen/microbiology , Spleen/pathology , Toll-Like Receptor 4/genetics
11.
J Biol Chem ; 288(32): 23064-74, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23792965

ABSTRACT

UDP-glucose dehydrogenase (Ugd) generates UDP-glucuronic acid, an important precursor for the production of many hexuronic acid-containing bacterial surface glycostructures. In Escherichia coli K-12, Ugd is important for biosynthesis of the environmentally regulated exopolysaccharide known as colanic acid, whereas in other E. coli isolates, the same enzyme is required for production of the constitutive group 1 capsular polysaccharides, which act as virulence determinants. Recent studies have implicated tyrosine phosphorylation in the activation of Ugd from E. coli K-12, although it is not known if this is a feature shared by bacterial Ugd proteins. The activities of Ugd from E. coli K-12 and from the group 1 capsule prototype (serotype K30) were compared. Surprisingly, for both enzymes, site-directed Tyr → Phe mutants affecting the previously proposed phosphorylation site retained similar kinetic properties to the wild-type protein. Purified Ugd from E. coli K-12 had significant levels of NAD substrate inhibition, which could be alleviated by the addition of ATP and several other nucleotide triphosphates. Mutations in a previously identified UDP-glucuronic acid allosteric binding site decreased the binding affinity of the nucleotide triphosphate. Ugd from E. coli serotype K30 was not inhibited by NAD, but its activity still increased in the presence of ATP.


Subject(s)
Adenosine Triphosphate , Escherichia coli K12/enzymology , Escherichia coli Proteins , NAD , Uridine Diphosphate Glucose Dehydrogenase , Virulence Factors , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Amino Acid Substitution , Escherichia coli K12/genetics , Escherichia coli K12/pathogenicity , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Kinetics , Mutagenesis, Site-Directed , Mutation, Missense , NAD/chemistry , NAD/genetics , NAD/metabolism , Polysaccharides/biosynthesis , Polysaccharides/chemistry , Polysaccharides/genetics , Uridine Diphosphate Glucose Dehydrogenase/antagonists & inhibitors , Uridine Diphosphate Glucose Dehydrogenase/chemistry , Uridine Diphosphate Glucose Dehydrogenase/genetics , Uridine Diphosphate Glucose Dehydrogenase/metabolism , Virulence Factors/chemistry , Virulence Factors/genetics , Virulence Factors/metabolism
12.
Mol Microbiol ; 87(5): 939-50, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23350972

ABSTRACT

Escherichia coli has been the leading model organism for many decades. It is a fundamental player in modern biology, facilitating the molecular biology revolution of the last century. The acceptance of E. coli as model organism is predicated primarily on the study of one E. coli lineage; E. coli K-12. However, the antecedents of today's laboratory strains have undergone extensive mutagenesis to create genetically tractable offspring but which resulted in loss of several genetic traits such as O antigen expression. Here we have repaired the wbbL locus, restoring the ability of E. coli K-12 strain MG1655 to express the O antigen. We demonstrate that O antigen production results in drastic alterations of many phenotypes and the density of the O antigen is critical for the observed phenotypes. Importantly, O antigen production enables laboratory strains of E. coli to enter the gut of the Caenorhabditis elegans worm and to kill C. elegans at rates similar to pathogenic bacterial species. We demonstrate C. elegans killing is a feature of other commensal E. coli. We show killing is associated with bacterial resistance to mechanical shear and persistence in the C. elegans gut. These results suggest C. elegans is not an effective model of human-pathogenic E. coli infectious disease.


Subject(s)
Caenorhabditis elegans/microbiology , Escherichia coli K12/metabolism , Escherichia coli K12/pathogenicity , O Antigens/biosynthesis , Animals , Escherichia coli K12/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , O Antigens/genetics
13.
Bioeng Bugs ; 3(2): 133-7, 2012.
Article in English | MEDLINE | ID: mdl-22539026

ABSTRACT

The status of E. coli K-12 as an exclusively non-invasive, non-pathogenic bacterium has almost been incontrovertible. Our recent finding that a mutation in one of its main architectural protein, HU, converts E. coli K-12 to an actively invasive form suggests that gaining host cell entry might be an expedient survival tactic for traditional commensals during certain altered host conditions. The mutant E. coli (SK3842) exhibits properties usually associated with pathogenic bacteria: host cell invasion, phagosomal disruption and intracellular replication. However, unlike the situation with some pathogens, internalized SK3842 imparts anti-apoptotic and cyto-protective effects rather than lethality on the host cell, both in vitro and in vivo. Here, we show that SK3842 also provides colonization resistance against other invasive pathogens--a trait not shared by the parental commensal strain. Thus, the altered lifestyle of SK3842 encompasses characteristics both from traditional pathogens as well as beneficial probiotic strains.


Subject(s)
Escherichia coli K12/classification , Escherichia coli K12/growth & development , Apoptosis/drug effects , Epithelial Cells/microbiology , Epithelial Cells/pathology , Escherichia coli K12/pathogenicity , Host-Pathogen Interactions , Humans , Probiotics/metabolism
14.
Infect Immun ; 80(3): 1050-64, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22158741

ABSTRACT

The Yersinia pseudotuberculosis Ifp and InvC molecules are putative autotransporter proteins with a high homology to the invasin (InvA) protein. To characterize the function of these surface proteins, we expressed both factors in Escherichia coli K-12 and demonstrated the attachment of Ifp- and InvC-expressing bacteria to human-, mouse-, and pig-derived intestinal epithelial cells. Ifp also was found to mediate microcolony formation and internalization into polarized human enterocytes. The ifp and invC genes were not expressed under in vitro conditions but were found to be induced in the Peyer's patches of the mouse intestinal tract. In a murine coinfection model, the colonization of the Peyer's patches and the mesenteric lymph nodes of mice by the ifp-deficient strain was significantly reduced, and considerably fewer bacteria reached liver and spleen. The absence of InvC did not have a severe influence on bacterial colonization in the murine infection model, and it resulted in only a slightly reduced number of invC mutants in the Peyer's patches. The analysis of the host immune response demonstrated that the presence of Ifp and InvC reduced the recruitment of professional phagocytes, especially neutrophils, in the Peyer's patches. These findings support a role for the adhesins in modulating host-pathogen interactions that are important for immune defense.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion , Epithelial Cells/microbiology , Membrane Transport Proteins/metabolism , Virulence Factors/metabolism , Yersinia pseudotuberculosis/pathogenicity , Adhesins, Bacterial/genetics , Animals , Cell Line , Disease Models, Animal , Escherichia coli K12/genetics , Escherichia coli K12/pathogenicity , Female , Gene Expression , Humans , Intestines/microbiology , Lymph Nodes/microbiology , Membrane Transport Proteins/genetics , Mice , Mice, Inbred BALB C , Peyer's Patches/microbiology , Virulence , Virulence Factors/genetics , Yersinia pseudotuberculosis Infections/microbiology
15.
Infect Immun ; 80(1): 321-32, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21930758

ABSTRACT

Uropathogenic Escherichia coli (UPEC) is the primary cause of urinary tract infection (UTI) in the developed world. The major factors associated with virulence of UPEC are fimbrial adhesins, which mediate specific attachment to host receptors and trigger innate host responses. Another group of adhesins is represented by the autotransporter (AT) subgroup of proteins. The genome-sequenced prototype UPEC strain CFT073 contains 11 putative AT-encoding genes. In this study, we have performed a detailed molecular characterization of two closely related AT adhesins from CFT073: UpaB (c0426) and UpaC (c0478). PCR screening revealed that the upaB and upaC AT-encoding genes are common in E. coli. The upaB and upaC genes were cloned and characterized in a recombinant E. coli K-12 strain background. This revealed that they encode proteins located at the cell surface but possess different functional properties: UpaB mediates adherence to several ECM proteins, while UpaC expression is associated with increased biofilm formation. In CFT073, upaB is expressed while upaC is transcriptionally repressed by the global regulator H-NS. In competitive colonization experiments employing the mouse UTI model, CFT073 significantly outcompeted its upaB (but not upaC) isogenic mutant strain in the bladder. This attenuated phenotype was also observed in single-challenge experiments, where deletion of the upaB gene in CFT073 significantly reduced early colonization of the bladder.


Subject(s)
Adhesins, Bacterial/metabolism , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/metabolism , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/pathogenicity , Virulence Factors/metabolism , Adhesins, Bacterial/genetics , Animals , Biofilms/growth & development , Cloning, Molecular , DNA, Bacterial/genetics , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Escherichia coli K12/genetics , Escherichia coli K12/pathogenicity , Escherichia coli Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Fimbriae Proteins/metabolism , Gene Expression Profiling , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Urinary Tract Infections/microbiology , Urinary Tract Infections/pathology , Virulence Factors/genetics
16.
mBio ; 2(5)2011.
Article in English | MEDLINE | ID: mdl-21896677

ABSTRACT

UNLABELLED: The HUα(E38K, V42L) mutant of the bacterial histone-like protein HU causes a major change in the transcription profile of the commensal organism Escherichia coli K-12 (Kar S, Edgar R, Adhya S, Proc. Natl. Acad. Sci. U. S. A. 102:16397-16402, 2005). Among the upregulated genes are several related to pathogenic interactions with mammalian cells, as evidenced by the expression of curli fibers, Ivy, and hemolysin E. When E. coli K-12/ HUα(E38K, V42L) was added to Int-407 cells, there was host cell invasion, phagosomal disruption, and intracellular replication. The invasive trait was also retained in a murine ileal loop model and intestinal explant assays. In addition to invasion, the internalized bacteria caused a novel subversion of host cell apoptosis through modification and regulation of the BH3-only proteins Bim(EL) and Puma. Changes in the transcription profile were attributed to positive supercoiling of DNA leading to the altered availability of relevant promoters. Using the E. coli K-12/HUα(E38K, V42L) variant as a model, we propose that traditional commensal E. coli can adopt an invasive lifestyle through reprogramming its cellular transcription, without gross genetic changes. IMPORTANCE: Escherichia coli K-12 is well established as a benign laboratory strain and a human intestinal commensal. Recent evidences, however, indicate that the typical noninvasive nature of resident E. coli can be reversed under specific circumstances even in the absence of any major genomic flux. We previously engineered an E. coli strain with a mutant histone-like protein, HU, which exhibited significant changes in nucleoid organization and global transcription. Here we showed that the changes induced by the mutant HU have critical functional consequences: from a strict extracellular existence, the mutant E. coli adopts an almost obligate intracellular lifestyle. The internalized E. coli exhibits many of the prototypical characteristics of traditional intracellular bacteria, like phagosomal escape, intracellular replication, and subversion of host cell apoptosis. We suggest that E. coli K-12 can switch between widely divergent lifestyles in relation to mammalian host cells by reprogramming its cellular transcription program and without gross changes in its genomic content.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Escherichia coli K12/genetics , Escherichia coli K12/pathogenicity , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Mutation, Missense , Animals , Apoptosis , Cell Line , Disease Models, Animal , Epithelial Cells/microbiology , Female , Gene Expression , Humans , Ileum/microbiology , Mice , Mice, Inbred BALB C , Virulence
17.
J Biol Chem ; 286(37): 32026-35, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21768111

ABSTRACT

We have demonstrated that SlyA activates fimB expression and hence type 1 fimbriation, a virulence factor in Escherichia coli. SlyA is shown to bind to two operator sites (O(SA1) and O(SA2)), situated between 194 and 167 base pairs upstream of the fimB transcriptional start site. fimB expression is derepressed in an hns mutant and diminished by a slyA mutation in the presence of H-NS only. H-NS binds to multiple sites in the promoter region, including two sites (H-NS2 and H-NS3) that overlap O(SA1) and O(SA2), respectively. Mutations that disrupt either O(SA1) or O(SA2) eliminate or reduce the activating effect of SlyA but have different effects on the level of expression. We interpret these results as reflecting the relative competition between SlyA and H-NS binding. Moreover we show that SlyA is capable of displacing H-NS from its binding sites in vitro. We suggest SlyA binding prevents H-NS binding to H-NS2 and H-NS3 and the subsequent oligomerization of H-NS necessary for full inhibition of fimB expression. In addition, we show that SlyA activates fimB expression independently of two other known regulators of fimB expression, NanR and NagC. It is demonstrated that the rarely used UUG initiation codon limits slyA expression and that low SlyA levels limit fimB expression. Furthermore, Western blot analysis shows that cells grown in rich-defined medium contain ~1000 SlyA dimers per cell whereas those grown in minimal medium contain >20% more SlyA. This study extends our understanding of the role that SlyA plays in the host-bacterial relationship.


Subject(s)
Bacterial Proteins/metabolism , DNA-Binding Proteins/biosynthesis , Escherichia coli K12/metabolism , Escherichia coli Proteins/biosynthesis , Gene Expression Regulation, Bacterial , Integrases/biosynthesis , Transcription Factors/metabolism , Virulence Factors/biosynthesis , Bacterial Proteins/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Escherichia coli K12/genetics , Escherichia coli K12/pathogenicity , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Host-Pathogen Interactions/genetics , Integrases/genetics , Mutation , Operator Regions, Genetic/genetics , Protein Binding/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Virulence Factors/genetics
18.
J Bacteriol ; 193(18): 4813-20, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21764925

ABSTRACT

Heat-resistant agglutinin 1 (Hra1) is an accessory colonization factor of enteroaggregative Escherichia coli (EAEC) strain 042. Tia, a close homolog of Hra1, is an invasin and adhesin that has been described in enterotoxigenic E. coli. We devised a PCR-restriction fragment length polymorphism screen for the associated genes and found that they occur among 55 (36.7%) of the enteroaggregative E. coli isolates screened, as well as lower proportions of enterotoxigenic, enteropathogenic, enterohemorrhagic, and commensal E. coli isolates. Overall, 25%, 8%, and 3% of 150 EAEC strains harbored hra1 alone, tia alone, or both genes, respectively. One EAEC isolate, 60A, produced an amplicon with a unique restriction profile, distinct from those of hra1 and tia. We cloned and sequenced the full-length agglutinin gene from strain 60A and have designated it hra2. The hra2 gene was not detected in any of 257 diarrheagenic E. coli isolates in our collection but is present in the genome of Salmonella enterica serovar Heidelberg strain SL476. The cloned hra2 gene from strain 60A, which encodes a predicted amino acid sequence that is 64% identical to that of Hra1 and 68% identical to that of Tia, was sufficient to confer adherence on E. coli K-12. We constructed an hra2 deletion mutant of EAEC strain 60A. The mutant was deficient in adherence but not autoaggregation or invasion, pointing to a functional distinction from the autoagglutinin Hra1 and the Tia invasin. Hra1, Tia, and the novel accessory adhesin Hra2 are members of a family of integral outer membrane proteins that confer different colonization-associated phenotypes.


Subject(s)
Adhesins, Escherichia coli/metabolism , Agglutinins/metabolism , Bacterial Adhesion , Escherichia coli/pathogenicity , Adhesins, Escherichia coli/genetics , Agglutinins/genetics , Cloning, Molecular , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli K12/genetics , Escherichia coli K12/pathogenicity , Gene Deletion , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Salmonella enterica/genetics , Sequence Analysis, DNA , Sequence Homology, Amino Acid
19.
Dev Comp Immunol ; 35(10): 1091-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21527277

ABSTRACT

To better understand the molecular basis underlying aphid immune tolerance to beneficial bacteria and immune defense to pathogenic bacteria, we characterized how the pea aphid Acyrthosiphon pisum responds to Escherichia coli K-12 infections. E. coli bacteria, usually cleared in the hemolymph of other insect species, were capable of growing exponentially and killing aphids within a few days. Red fluorescence protein expressing E. coli K-12 laboratory strain multiplied in the aphid hemolymph as well as in the digestive tract, resulting in death of infected aphids. Selected gene deletion mutants of the E. coli K-12 predicted to have reduced virulence during systemic infections showed no difference in either replication or killing rate when compared to the wild type E. coli strain. Of note, however, the XL1-Blue E. coli K-12 strain exhibited a significant lag phase before multiplying and killing aphids. This bacterial strain has recently been shown to be more sensitive to oxidative stress than other E. coli K-12 strains, revealing a potential role for reactive oxygen species-mediated defenses in the otherwise reduced aphid immune system.


Subject(s)
Aphids , Escherichia coli K12 , Host-Pathogen Interactions/immunology , Immunity, Innate/physiology , Reactive Oxygen Species/immunology , Animals , Aphids/genetics , Aphids/immunology , Aphids/microbiology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Escherichia coli Infections/microbiology , Escherichia coli K12/genetics , Escherichia coli K12/growth & development , Escherichia coli K12/pathogenicity , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Gastrointestinal Tract/microbiology , Hemolymph/microbiology , Membrane Proteins/genetics , Membrane Proteins/immunology , Porins/genetics , Porins/immunology , Protein Kinases/genetics , Protein Kinases/immunology , Sequence Deletion/immunology , Symbiosis/immunology
20.
Infect Immun ; 79(6): 2430-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21422176

ABSTRACT

We previously isolated a spontaneous mutant of Escherichia coli K-12, strain MG1655, following passage through the streptomycin-treated mouse intestine, that has colonization traits superior to the wild-type parent strain (M. P. Leatham et al., Infect. Immun. 73:8039-8049, 2005). This intestine-adapted strain (E. coli MG1655*) grew faster on several different carbon sources than the wild type and was nonmotile due to deletion of the flhD gene. We now report the results of several high-throughput genomic analysis approaches to further characterize E. coli MG1655*. Whole-genome pyrosequencing did not reveal any changes on its genome, aside from the deletion at the flhDC locus, that could explain the colonization advantage of E. coli MG1655*. Microarray analysis revealed modest yet significant induction of catabolic gene systems across the genome in both E. coli MG1655* and an isogenic flhD mutant constructed in the laboratory. Catabolome analysis with Biolog GN2 microplates revealed an enhanced ability of both E. coli MG1655* and the isogenic flhD mutant to oxidize a variety of carbon sources. The results show that intestine-adapted E. coli MG1655* is more fit than the wild type for intestinal colonization, because loss of FlhD results in elevated expression of genes involved in carbon and energy metabolism, resulting in more efficient carbon source utilization and a higher intestinal population. Hence, mutations that enhance metabolic efficiency confer a colonization advantage.


Subject(s)
Escherichia coli K12/pathogenicity , Intestines/microbiology , Animals , Base Sequence , Escherichia coli K12/genetics , Genes, Bacterial/genetics , Genes, Bacterial/physiology , Genome/genetics , Genome-Wide Association Study , Genotype , Male , Mice , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Phenotype , Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL