Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
PLoS One ; 9(9): e106005, 2014.
Article in English | MEDLINE | ID: mdl-25198673

ABSTRACT

BACKGROUND: We studied the expression of sulphated glycosaminoglycans (GAGs) in coeliac disease (CD) mucosa, as they are critical determinants of tissue volume, which increases in active disease. We also examined mucosal expression of IL-6, which stimulates excess GAG synthesis in disorders such as Grave's ophthalmopathy. METHODS: We stained archival jejunal biopsies from 5 children with CD at diagnosis, on gluten-free diet and challenge for sulphated GAGs. We then examined duodenal biopsies from 9 children with CD compared to 9 histological normal controls, staining for sulphated GAGs, heparan sulphate proteoglycans (HSPG), short-chain HSPG (Δ-HSPG) and the proteoglycan syndecan-1 (CD138), which is expressed on epithelium and plasma cells. We confirmed findings with a second monoclonal in another 12 coeliac children. We determined mucosal IL-6 expression by immunohistochemistry and PCR in 9 further cases and controls, and used quantitative real time PCR for other Th17 pathway cytokines in an additional 10 cases and controls. RESULTS: In CD, HSPG expression was lost in the epithelial compartment but contrastingly maintained within an expanded lamina propria. Within the upper lamina propria, clusters of syndecan-1(+) plasma cells formed extensive syncytial sheets, comprising adherent plasma cells, lysed cells with punctate cytoplasmic staining and shed syndecan ectodomains. A dense infiltrate of IL-6(+) mononuclear cells was detected in active coeliac disease, also localised to the upper lamina propria, with significantly increased mRNA expression of IL-6 and IL-17A but not IL-23 p19. CONCLUSIONS: Matrix expansion, through syndecan-1(+) cell recruitment and lamina propria GAG increase, underpins villous atrophy in coeliac disease. The syndecan-1(+) cell syncytia and excess GAG production recapitulate elements of the invertebrate encapsulation reaction, itself dependent on insect transglutaminase and glutaminated early response proteins. As in other matrix expansion disorders, IL-6 is upregulated and represents a logical target for immunotherapy in patients with coeliac disease refractory to gluten-free diet.


Subject(s)
Celiac Disease/metabolism , Extracellular Matrix/metabolism , Giant Cells/metabolism , Intestinal Mucosa/pathology , Syndecan-1/metabolism , Adolescent , Base Sequence , Biopsy , Celiac Disease/diagnosis , Celiac Disease/pathology , Child , Child, Preschool , DNA Primers , Glycosaminoglycans/metabolism , Humans , Interleukin-6/genetics , Intestinal Mucosa/metabolism , Real-Time Polymerase Chain Reaction
2.
Cell Microbiol ; 16(8): 1255-66, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24612002

ABSTRACT

Haemolytic uraemic syndrome caused by Shiga toxin-producing E. coli (STEC) is dependent on release of Shiga toxins (Stxs) during intestinal infection and subsequent absorption into the bloodstream. An understanding of Stx-related events in the human gut is limited due to lack of suitable experimental models. In this study, we have used a vertical diffusion chamber system with polarized human colon carcinoma cells to simulate the microaerobic (MA) environment in the human intestine and investigate its influence on Stx release and translocation during STEC O157:H7 and O104:H4 infection. Stx2 was the major toxin type released during infection. Whereas microaerobiosis significantly reduced bacterial growth as well as Stx production and release into the medium, Stx translocation across the epithelial monolayer was enhanced under MA versus aerobic conditions. Increased Stx transport was dependent on STEC infection and occurred via a transcellular pathway other than macropinocytosis. While MA conditions had a similar general effect on Stx release and absorption during infection with STEC O157:H7 and O104:H4, both serotypes showed considerable differences in colonization, Stx production, and Stx translocation which suggest alternative virulence strategies. Taken together, our study suggests that the MA environment in the human colon may modulate Stx-related events and enhance Stx absorption during STEC infection.


Subject(s)
Colonic Diseases/pathology , Escherichia coli Infections/pathology , Shiga Toxin/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity , Anaerobiosis , Animals , Cell Line, Tumor , Chlorocebus aethiops , Colonic Diseases/microbiology , Cytochalasin D/pharmacology , Escherichia coli Infections/microbiology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/pathology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Oxygen , Pinocytosis/drug effects , Shiga Toxin/biosynthesis , Shiga-Toxigenic Escherichia coli/classification , Vero Cells
3.
Inflamm Bowel Dis ; 18(4): 624-40, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21887731

ABSTRACT

BACKGROUND: Enterotoxin-producing Staphylococcus aureus may cause severe inflammatory intestinal disease, particularly in infants or immunodeficient or elderly patients. They are also recognized to be associated with sudden infant death syndrome. Little is known, however, about mucosal responses to staphylococci. METHODS: The mucosal lesion in three infants with staphylococcal enterocolitis was assessed by immunohistochemistry and electron microscopy. The organisms underwent extensive molecular analysis. Their toxins were assessed for capacity to induce T-cell activation and host mucosal responses examined by in vitro organ culture. Epithelial responses were studied by coculture with HEp-2 and Caco-2 cells. RESULTS: Intestinal biopsies from the patients showed marked epithelial damage with mucosal inflammation. The three staphylococci, representing two distinct clones, were methicillin-sensitive, producing SEG/I enterotoxins and Rho-inactivating EDIN toxins. Their enterotoxins potently activated T cells, but only whole organisms could induce in vitro enteropathy, characterized by remarkable epithelial desquamation uninhibited by tacrolimus. EDIN-producing staphylococci, but not their supernatants, induced striking cytopathy in HEp-2 epithelial cells but not in Caco-2 cells. Although HEp-2 and Caco-2 cells produced similar IL-8, CCL20, and cathelicidin LL37 responses upon bacterial exposure, only Caco-2 cells expressed mRNA for the ß-defensins HBD2 and HBD3, while HEp-2 cells were unable to do so. CONCLUSIONS: Staphylococci induce enterocolitis by a combination of direct enterocyte cytopathy mediated by EDIN toxins, disrupting the epithelial barrier, and enterotoxin superantigen-induced mucosal T-cell activation. Gut epithelial production of ß-defensins may contribute to host defense against invasive staphylococcal disease.


Subject(s)
Enterocolitis/immunology , Lymphocyte Activation/immunology , Staphylococcal Infections/immunology , Superantigens/immunology , T-Lymphocytes/immunology , Biopsy , Cell Line , Coculture Techniques , Enterocolitis/microbiology , Enterocolitis/pathology , Enterotoxins/immunology , Enterotoxins/toxicity , Female , Humans , Infant , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Staphylococcal Infections/pathology , Staphylococcus aureus/immunology , Staphylococcus aureus/ultrastructure , Tacrolimus , beta-Defensins/biosynthesis
4.
PLoS One ; 6(10): e27030, 2011.
Article in English | MEDLINE | ID: mdl-22046438

ABSTRACT

BACKGROUND: Enteropathogenic (EPEC) and Enteroaggregative (EAEC) E. coli have similar, but distinct clinical symptoms and modes of pathogenesis. Nevertheless when they infect the gastrointestinal tract, it is thought that their flagellin causes IL-8 release leading to neutrophil recruitment and gastroenteritis. However, this may not be the whole story as the effect of bacterial adherence to IEC innate response(s) remains unclear. Therefore, we have characterized which bacterial motifs contribute to the innate epithelial response to EPEC and EAEC, using a range of EPEC and EAEC isogenic mutant strains. METHODOLOGY: Caco-2 and HEp-2 cell lines were exposed to prototypical EPEC strain E2348/69 or EAEC strain O42, in addition to a range of isogenic mutant strains. E69 [LPS, non-motile, non-adherent, type three secretion system (TTSS) negative, signalling negative] or O42 [non-motile, non-adherent]. IL-8 and CCL20 protein secretion was measured. Bacterial surface structures were assessed by negative staining Transmission Electron Microscopy. The Fluorescent-actin staining test was carried out to determine bacterial adherence. RESULTS: Previous studies have reported a balance between the host pro-inflammatory response and microbial suppression of this response. In our system an overall balance towards the host pro-inflammatory response is seen with the E69 WT and to a greater extent O42 WT, which is in fit with clinical symptoms. On removal of the external EPEC structures flagella, LPS, BFP, EspA and EspC; and EAEC flagella and AAF, the host inflammatory response is reduced. However, removal of E69 lymphostatin increases the host inflammatory response suggesting involvement in the bacterial mediated anti-inflammatory response. CONCLUSION: Epithelial responses were due to combinations of bacterial agonists, with host-bacterial contact a key determinant of these innate responses. Host epithelial recognition was offset by the microbe's ability to down-regulate the inflammatory response. Understanding the complexity of this host-microbial balance will contribute to improved vaccine design for infectious gastroenteritis.


Subject(s)
Bacterial Adhesion/immunology , Enteropathogenic Escherichia coli/immunology , Epithelial Cells/immunology , Escherichia coli/immunology , Immunity, Innate , Animals , Cell Line , Host-Pathogen Interactions/immunology , Inflammation/microbiology
5.
J Pediatr Gastroenterol Nutr ; 53(1): 48-54, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21694535

ABSTRACT

OBJECTIVES: Autosomal recessive, congenital chloride diarrhea (CLD) is a form of persistent secretory diarrhea, presenting with polyhydramnios and intractable diarrhea from birth. CLD is caused by mutations in the SLC26A3 gene, encoding a Na+-independent Cl/HCO3- exchanger. The diagnosis is generally made on the basis of high fecal chloride concentration in patients with serum electrolyte homoeostasis corrected by salt substitution. We aimed to evaluate the role of diagnostic genetic testing in CLD. PATIENTS AND METHODS: Clinical and laboratory data were collected from 8 unrelated children diagnosed as having or suspected to have CLD. The evaluation included physical examination, routine clinical chemistry, and SLC26A3 mutation analysis by direct sequencing of DNA extracted from buccal swabs or peripheral leukocytes. RESULTS: CLD was initially diagnosed on high fecal chloride concentrations in 7 patients, and by mutation analysis in 1 patient. In 3 of these patients the correct diagnosis was made more than 6 months after birth. We identified SLC26A3 mutations on both alleles in all 8 patients with CLD, including 3 novel missense and 4 novel truncating mutations. We present a compilation of reported SLC26A3 mutations and polymorphisms. CONCLUSIONS: The diagnosis and therapy of CLD were considerably delayed in 3 of 8 patients from this series, highlighting the potential of misdiagnosing CLD. We add 7 novel mutations, including 3 missense changes of highly conserved residues to a total of 41 mutations in this gene. Molecular analysis is efficient and should be considered as a means of early diagnosis of CLD, especially if the clinical diagnosis remains uncertain.


Subject(s)
Chloride-Bicarbonate Antiporters/genetics , Diarrhea/congenital , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/genetics , Molecular Diagnostic Techniques , Mutant Proteins/genetics , Amino Acid Sequence , Child , Child, Preschool , Chloride-Bicarbonate Antiporters/chemistry , Chlorides/analysis , DNA Mutational Analysis , Databases, Protein , Delayed Diagnosis/prevention & control , Diarrhea/diagnosis , Diarrhea/genetics , Feces/chemistry , Female , Humans , Infant , Infant, Newborn , Male , Molecular Sequence Data , Mutant Proteins/chemistry , Mutation, Missense , Polymorphism, Genetic , Sequence Alignment , Sulfate Transporters
6.
PLoS One ; 5(10): e15389, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-21042595

ABSTRACT

BACKGROUND: Human beta-defensins (hBDs) are antimicrobial peptides known to play a major role in intestinal innate host defence. Altered mucosal expression of hBDs has been suggested to be implicated in chronic inflammatory bowel disease pathogenesis. However, little is known about expression of these peptides in children. METHODS: Intestinal biopsies were obtained from the duodenum (n = 88), terminal ileum (n = 90) and ascending colon (n = 105) of children with Crohn's disease (n = 26), ulcerative colitis (n = 11) and healthy controls (n = 16). Quantitative real-time (RT) PCR was performed and absolute mRNA copy numbers analyzed for hBD1-3 as well as inflammatory cytokines IL-8 and TNF-alpha. RESULTS: Significant induction of hBD2 and hBD3 was observed in the inflamed terminal ileum and ascending colon of IBD children. In the ascending colon induction of hBD2 was found to be significantly lower in children with Crohn's disease compared to ulcerative colitis. A strong correlation was found between inducible defensins hBD2 and 3 and the inflammatory cytokines IL-8 and TNF-alpha, both in the terminal ileum and ascending colon. CONCLUSION: Our study demonstrates distinct changes in hBD expression throughout the intestinal tract of children with IBD, lending further support for their potential role in disease pathogenesis.


Subject(s)
Inflammatory Bowel Diseases/metabolism , beta-Defensins/metabolism , Case-Control Studies , Child , Chronic Disease , Cytokines/metabolism , Humans
7.
PLoS Pathog ; 6(5): e1000898, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20485572

ABSTRACT

Many bacterial pathogens utilize a type III secretion system to deliver multiple effector proteins into host cells. Here we found that the type III effectors, NleE from enteropathogenic E. coli (EPEC) and OspZ from Shigella, blocked translocation of the p65 subunit of the transcription factor, NF-kappaB, to the host cell nucleus. NF-kappaB inhibition by NleE was associated with decreased IL-8 expression in EPEC-infected intestinal epithelial cells. Ectopically expressed NleE also blocked nuclear translocation of p65 and c-Rel, but not p50 or STAT1/2. NleE homologues from other attaching and effacing pathogens as well OspZ from Shigella flexneri 6 and Shigella boydii, also inhibited NF-kappaB activation and p65 nuclear import; however, a truncated form of OspZ from S. flexneri 2a that carries a 36 amino acid deletion at the C-terminus had no inhibitory activity. We determined that the C-termini of NleE and full length OspZ were functionally interchangeable and identified a six amino acid motif, IDSY(M/I)K, that was important for both NleE- and OspZ-mediated inhibition of NF-kappaB activity. We also established that NleB, encoded directly upstream from NleE, suppressed NF-kappaB activation. Whereas NleE inhibited both TNFalpha and IL-1beta stimulated p65 nuclear translocation and IkappaB degradation, NleB inhibited the TNFalpha pathway only. Neither NleE nor NleB inhibited AP-1 activation, suggesting that the modulatory activity of the effectors was specific for NF-kappaB signaling. Overall our data show that EPEC and Shigella have evolved similar T3SS-dependent means to manipulate host inflammatory pathways by interfering with the activation of selected host transcriptional regulators.


Subject(s)
Escherichia coli O157/metabolism , Escherichia coli Proteins/metabolism , Shigella boydii/metabolism , Shigella flexneri/metabolism , Transcription Factor RelA/metabolism , Virulence Factors/metabolism , Active Transport, Cell Nucleus/physiology , Caco-2 Cells , Dysentery, Bacillary/immunology , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/microbiology , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli O157/pathogenicity , HeLa Cells , Humans , I-kappa B Proteins/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , NF-kappa B p50 Subunit/metabolism , Proto-Oncogene Proteins c-rel/metabolism , RNA, Messenger/metabolism , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Shigella boydii/pathogenicity , Shigella flexneri/pathogenicity , Transcriptional Activation/physiology , Virulence
8.
Environ Microbiol ; 12(9): 2426-35, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20406285

ABSTRACT

Advances in the understanding of the pathogenesis of enterohaemorrhagic Escherichia coli (EHEC) have greatly benefited from the use of human epithelial cell lines under aerobic conditions. However, in the target site of EHEC infection, the human intestine, conditions are microaerobic. In our study we used polarized human colon carcinoma cells in a vertical diffusion chamber system to investigate the influence of reduced apical oxygen levels on EHEC colonization. While apical microaerobiosis did not affect cell integrity and barrier function, numbers of adherent bacteria were significantly increased under low compared with high apical oxygen concentrations. In addition, expression and translocation of EHEC type III secreted (T3S) effector proteins was considerably enhanced under microaerobic conditions and dependent on the presence of host cells. Increased colonization was mainly mediated via EspA as adherence levels of an isogenic deletion mutant were not influenced by low oxygen levels. Other potential adherence factors (E. coli common pilus and flagella) were only minimally expressed under high and low oxygen levels. Addition of nitrate and trimethylamine N-oxide as terminal electron acceptors for anaerobic respiration failed to further increase bacterial colonization or T3S under microaerobiosis. This study indicates that EHEC T3S and colonization are enhanced by the microaerobic environment in the gut and therefore might be underestimated in conventional aerobic cell culture systems.


Subject(s)
Bacterial Adhesion , Enterohemorrhagic Escherichia coli/growth & development , Epithelial Cells/microbiology , Escherichia coli Infections/microbiology , Aerobiosis , Cell Line, Tumor , Cell Polarity , Culture Media/chemistry , Enterohemorrhagic Escherichia coli/metabolism , Epithelial Cells/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Fimbriae, Bacterial/metabolism , Humans , Oxygen/metabolism
9.
Mol Microbiol ; 75(2): 308-23, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19889090

ABSTRACT

Attaching and effacing (A/E) lesions and actin polymerization, the hallmark of enteropathogenic Escherichia coli (EPEC), enterohemorrhagic E. coli (EHEC) and Citrobacter rodentium (CR) infections, are dependent on the effector Tir. Phosphorylation of Tir(EPEC/CR) Y474/1 leads to recruitment of Nck and neural Wiskott-Aldrich syndrome protein (N-WASP) and strong actin polymerization in cultured cells. Tir(EPEC/CR) also contains an Asn-Pro-Tyr (NPY(454/1)) motif, which triggers weak actin polymerization. In EHEC the NPY(458) actin polymerization pathway is amplified by TccP/EspF(U), which is recruited to Tir via IRSp53 and/or insulin receptor tyrosine kinase substrate (IRTKS). Here we used C. rodentium to investigate the different Tir signalling pathways in vivo. Following infection with wild-type C. rodentium IRTKS, but not IRSp53, was recruited to the bacterial attachment sites. Similar results were seen after infection of human ileal explants with EHEC. Mutating Y471 or Y451 in Tir(CR) abolished recruitment of Nck and IRTKS respectively, but did not affect recruitment of N-WASP or A/E lesion formation. This suggests that despite their crucial role in actin polymerization in cultured cells the Tir:Nck and Tir:IRTKS pathways are not essential for N-WASP recruitment or A/E lesion formation in vivo. Importantly, wild-type C. rodentium out-competed the tir tyrosine mutants during mixed infections. These results uncouple the Tir:Nck and Tir:IRTKS pathways from A/E lesion formation in vivo but assign them an important in vivo role.


Subject(s)
Enterohemorrhagic Escherichia coli/physiology , Enteropathogenic Escherichia coli/physiology , Escherichia coli Infections/physiopathology , 3T3 Cells/microbiology , Actins/metabolism , Animals , Bacterial Adhesion , Binding Sites , Cells, Cultured , Citrobacter rodentium/genetics , Citrobacter rodentium/pathogenicity , Citrobacter rodentium/physiology , Enterobacteriaceae Infections/physiopathology , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/pathogenicity , Enteropathogenic Escherichia coli/pathogenicity , Humans , Ileum/microbiology , Mice , Mutagenesis , Peptides/genetics , Signal Transduction , Tyrosine/genetics , Wiskott-Aldrich Syndrome/physiopathology , Wiskott-Aldrich Syndrome Protein/physiology , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics
10.
J Bacteriol ; 191(11): 3451-61, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19218393

ABSTRACT

Although the bundle-forming pilus (BFP) of enteropathogenic Escherichia coli (EPEC) mediates microcolony formation on epithelial cells, the adherence of BFP-deficient mutants is significantly abrogated, but the mutants are still adherent due to the presence of intimin and possibly other adhesins. In this study we investigated the contribution of the recently described E. coli common pilus (ECP) to the overall adherence properties of EPEC. We found that ECP and BFP structures can be simultaneously observed in the course (between zero time and 7 h during infection) of formation of localized adherence on cultured epithelial cells. These two pilus types colocalized at different levels of the microcolony topology, tethering the adhering bacteria. No evidence of BFP disappearance was found after prolonged infection. When expressed from a plasmid present in nonadherent E. coli HB101, ECP rendered this organism highly adherent at levels comparable to those of HB101 expressing the BFP. Purified ECP bound in a dose-dependent manner to epithelial cells, and the binding was blocked with anti-ECP antibodies, confirming that the pili possess adhesin properties. An ECP mutant showed only a modest reduction in adherence to cultured cells due to background expression levels of BFP and intimin. However, isogenic mutants not expressing EspA or BFP were significantly less adherent when the ecpA gene was also deleted. Furthermore, a DeltaespA DeltaecpA double mutant (unable to translocate Tir and to establish intimate adhesion) was at least 10-fold less adherent than the DeltaespA and DeltaecpA single mutants, even in the presence of BFP. A Delta bfp DeltaespA DeltaecpA triple mutant showed the least adherence compared to the wild type and all the isogenic mutant strains tested, suggesting that ECP plays a synergistic role in adherence. Our data indicate that ECP is an accessory factor that, in association with BFP and other adhesins, contributes to the multifactorial complex interaction of EPEC with host epithelial cells.


Subject(s)
Bacterial Adhesion/physiology , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Proteins/physiology , Fimbriae Proteins/physiology , Fimbriae, Bacterial/metabolism , Bacterial Adhesion/genetics , Enteropathogenic Escherichia coli/genetics , Enteropathogenic Escherichia coli/ultrastructure , Epithelial Cells/microbiology , Escherichia coli Proteins/genetics , Fimbriae Proteins/genetics , Fimbriae, Bacterial/genetics , Flow Cytometry , HT29 Cells , HeLa Cells , Humans , Microscopy, Electron, Transmission
11.
Environ Microbiol ; 11(4): 992-1006, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19187284

ABSTRACT

Curli are adhesive fimbriae of Escherichia coli and Salmonella enterica. Expression of curli (csgA) and cellulose (bcsA) is co-activated by the transcriptional activator CsgD. In this study, we investigated the contribution of curli and cellulose to the adhesive properties of enterohaemorragic (EHEC) O157:H7 and enteropathogenic E. coli (EPEC) O127:H6. While single mutations in csgA, csgD or bcsA in EPEC and EHEC had no dramatic effect on cell adherence, double csgAbcsA mutants were significantly less adherent than the single mutants or wild-type strains to human colonic HT-29 epithelial cells or to cow colon tissue in vitro. Overexpression of csgD (carried on plasmid pCP994) in a csgD mutant, but not in the single csgA or bscA mutants, led to significant increase in adherence and biofilm formation in EPEC and EHEC, suggesting that synchronized over-production of curli and cellulose enhances bacterial adherence. In line with this finding, csgD transcription was activated significantly in the presence of cultured epithelial cells as compared with growth in tissue culture medium. Analysis of the influence of virulence and global regulators in the production of curli in EPEC identified Fis (factor for inversion stimulation) as a, heretofore unrecognized, negative transcriptional regulator of csgA expression. An EPEC E2348/69Deltafis produced abundant amounts of curli whereas a double fis/csgD mutant yielded no detectable curli production. Our data suggest that curli and cellulose act in concert to favour host colonization, biofilm formation and survival in different environments.


Subject(s)
Bacterial Adhesion , Biofilms/growth & development , Cellulose/metabolism , Enteropathogenic Escherichia coli/physiology , Escherichia coli O157/physiology , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/physiology , Factor For Inversion Stimulation Protein/physiology , Animals , Cattle , Cell Line , Cellulose/genetics , Enteropathogenic Escherichia coli/genetics , Epithelial Cells/microbiology , Escherichia coli O157/genetics , Escherichia coli Proteins/genetics , Factor For Inversion Stimulation Protein/genetics , Gene Deletion , Gene Dosage , Gene Expression Regulation, Bacterial , Humans , Repressor Proteins/physiology , Trans-Activators/genetics
12.
Cell Microbiol ; 11(3): 521-30, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19134113

ABSTRACT

In vitro organ culture (IVOC) represents a gold standard model to study enteropathogenic E. coli (EPEC) infection of human intestinal mucosa. However, the optimal examination of the bacterial-host cell interaction requires a directional epithelial exposure, without serosal or cut surface stimulation. A polarized IVOC system (pIVOC) was developed in order to overcome such limitations: apical EPEC infection produced negligible bacterial leakage via biopsy edges, resulted in enhanced colonization compared with standard IVOC, and showed evidence of bacterial detachment, as in natural rabbit EPEC infections. Examination of mucosal innate immune responses in pIVOC showed both interleukin (IL)-8 mRNA and protein levels were significantly increased after apical EPEC infection. Increased IL-8 levels mainly depended on flagellin expression as fliC-negative EPEC did not elicit a significant IL-8 response despite increased mucosal colonization compared with wild-type EPEC. In addition, apical application of purified flagella significantly increased IL-8 protein levels over non-infected controls. Immunofluorescence staining of EPEC-infected small intestinal biopsies revealed apical and basolateral distribution of Toll-like receptor (TLR) 5 on epithelium, suggesting that EPEC can trigger mucosal IL-8 responses by apical flagellin/TLR5 interaction ex vivo and does not require access to the basolateral membrane as postulated in cell culture models.


Subject(s)
Enteropathogenic Escherichia coli/growth & development , Intestinal Mucosa/microbiology , Enteropathogenic Escherichia coli/immunology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Flagellin , Gene Deletion , Gene Expression Profiling , Humans , Interleukin-8/biosynthesis , Intestinal Mucosa/pathology , Organ Culture Techniques/methods , Toll-Like Receptor 5/biosynthesis
13.
Infect Immun ; 76(11): 4804-13, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18725419

ABSTRACT

The human pathogen enterohemorrhagic Escherichia coli (EHEC) O157:H7 colonizes human and animal gut via formation of attaching and effacing lesions. EHEC strains use a type III secretion system to translocate a battery of effector proteins into the mammalian host cell, which subvert diverse signal transduction pathways implicated in actin dynamics, phagocytosis, and innate immunity. The genomes of sequenced EHEC O157:H7 strains contain two copies of the effector protein gene nleH, which share 49% sequence similarity with the gene for the Shigella effector OspG, recently implicated in inhibition of migration of the transcriptional regulator NF-kappaB to the nucleus. In this study we investigated the role of NleH during EHEC O157:H7 infection of calves and lambs. We found that while EHEC DeltanleH colonized the bovine gut more efficiently than the wild-type strain, in lambs the wild-type strain exhibited a competitive advantage over the mutant during mixed infection. Using the mouse pathogen Citrobacter rodentium, which shares many virulence factors with EHEC O157:H7, including NleH, we observed that the wild-type strain exhibited a competitive advantage over the mutant during mixed infection. We found no measurable differences in T-cell infiltration or hyperplasia in colons of mice inoculated with the wild-type or the nleH mutant strain. Using NF-kappaB reporter mice carrying a transgene containing a luciferase reporter driven by three NF-kappaB response elements, we found that NleH causes an increase in NF-kappaB activity in the colonic mucosa. Consistent with this, we found that the nleH mutant triggered a significantly lower tumor necrosis factor alpha response than the wild-type strain.


Subject(s)
Citrobacter rodentium/pathogenicity , Escherichia coli O157/pathogenicity , Intestinal Mucosa/microbiology , Virulence Factors/metabolism , Animals , Cattle , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/metabolism , Enterobacteriaceae Infections/pathology , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/pathology , Fluorescent Antibody Technique, Indirect , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , NF-kappa B/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Swine , Virulence Factors/genetics
14.
Infect Immun ; 76(10): 4669-76, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18678675

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) O157:H7 is an important human pathogen that colonizes the gut mucosa via attaching and effacing (A/E) lesions; A/E lesion formation in vivo and ex vivo is dependent on the type III secretion system (T3SS) effector Tir. Infection of cultured cells by EHEC leads to induction of localized actin polymerization, which is dependent on Tir and a second T3SS effector protein, TccP, also known as EspF(U). Recently, cortactin was shown to bind both the N terminus of Tir and TccP via its SH3 domain and to play a role in EHEC-triggered actin polymerization in vitro. In this study, we investigated the recruitment of cortactin to the site of EHEC adhesion during infection of in vitro-cultured cells and mucosal surfaces ex vivo (using human terminal ileal in vitro organ cultures [IVOC]). We have shown that cortactin is recruited to the site of EHEC adhesion in vitro downstream of TccP and N-WASP. Deletion of the entire N terminus of Tir or replacing the N-terminal polyproline region with alanines did not abrogate actin polymerization or cortactin recruitment. In contrast, recruitment of cortactin to the site of EHEC adhesion in IVOC is TccP independent. These results imply that cortactin is recruited to the site of EHEC adhesion in vitro and ex vivo by different mechanisms and suggest that cortactin might have a role during EHEC infection of mucosal surfaces.


Subject(s)
Bacterial Adhesion , Cortactin/metabolism , Escherichia coli O157/physiology , Actins/metabolism , Adolescent , Animals , Cell Line , Cells, Cultured , Child , Epithelial Cells/microbiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Intestinal Mucosa/microbiology , Mice , Organ Culture Techniques , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism
15.
FEMS Microbiol Lett ; 283(2): 196-202, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18422621

ABSTRACT

In this study, we used mouse ileal loops to investigate the interaction of enterohemorrhagic Escherichia coli (EHEC) O157:H7 with the mouse intestinal mucosa. With a dose of 10(9) and 3 h incubation, EHEC O157 was detected in the lumen and to a lesser extent associated with the epithelium. Typical attaching and effacing (A/E) lesions were seen, albeit infrequently. While the effector protein Tir was essential for A/E lesion formation, the bacterial type III secretion system adaptor protein TccP was dispensable. These results suggest that A/E lesions on mouse intestinal mucosa can be formed independently of robust actin polymerization.


Subject(s)
Bacterial Adhesion , Escherichia coli O157/physiology , Intestinal Mucosa/microbiology , Animals , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Ileum/microbiology , Mice , Mice, Inbred ICR , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
16.
Cell Microbiol ; 10(3): 549-56, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18053003

ABSTRACT

Enteropathogenic and enterohaemorrhagic Escherichia coli (EPEC and EHEC) colonize the gut mucosa via attaching and effacing (A/E) lesions. For years cultured cells were used as model systems to study A/E lesion formation, which showed actin accumulation under attached bacteria that can be raised above the plasma membrane in a pedestal-shaped structure. Studies of prototypical strains revealed that although both converge on N-WASP EPEC and EHEC O157:H7 use different actin polymerization pathways. While EPEC use the Tir-Nck pathway, Tir(EHECO157) cooperates with TccP/EspF(U) to activate N-WASP. However, recent in vitro studies revealed a common EPEC and EHEC Tir-dependent and Nck-independent inefficient actin polymerization pathway. Unexpectedly, bacterial populations studies demonstrated that most non-O157 EHEC strains and EPEC lineage 2 strains can utilize both the Nck and TccP2 pathways in vitro. Importantly, in vivo and ex vivo mucosal infections have shown efficient A/E lesion formation independently of Nck and TccP. This review covers the progression in our understanding of EPEC and EHEC infection, through the different milestones obtained using cultured cells, to the realization that EPEC and EHEC have much more in common than previously appreciated and that mucosal attachment and microvillous effacement may be the key events, rather than pedestal formation.


Subject(s)
Actins/metabolism , Bacterial Adhesion , Enterohemorrhagic Escherichia coli/physiology , Enteropathogenic Escherichia coli/physiology , Escherichia coli Proteins/metabolism , Receptors, Cell Surface/metabolism , Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Oncogene Proteins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism
17.
Infect Immun ; 76(1): 361-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17984209

ABSTRACT

Typical enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) employ either Nck, TccP/TccP2, or Nck and TccP/TccP2 pathways to activate the neuronal Wiskott-Aldrich syndrome protein (N-WASP) and to trigger actin polymerization in cultured cells. This phenotype is used as a marker for the pathogenic potential of EPEC and EHEC strains. In this paper we report that EPEC O125:H6, which represents a large category of strains, lacks the ability to utilize either Nck or TccP/TccP2 and hence triggers actin polymerization in vitro only inefficiently. However, we show that infection of human intestinal biopsies with EPEC O125:H6 results in formation of typical attaching and effacing lesions. Expression of TccP in EPEC O125:H6, which harbors an EHEC O157-like Tir, resulted in efficient actin polymerization in vitro and enhanced colonization of human intestinal in vitro organ cultures with detectable N-WASP and electron-dense material at the site of bacterial adhesion. These results show the existence of a natural category of EPEC that colonizes the gut mucosa using Nck- and TccP-independent mechanisms. Importantly, the results highlight yet again the fact that conclusions made on the basis of in vitro cell culture models cannot be extrapolated wholesale to infection of mucosal surfaces and that the ability to induce actin polymerization on cultured cells should not be used as a definitive marker for EPEC and EHEC virulence.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Enteropathogenic Escherichia coli/genetics , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Intestines/pathology , Oncogene Proteins/metabolism , Actins/metabolism , Bacterial Adhesion , Biopsy , Escherichia coli Proteins/genetics , Gene Expression Regulation , HeLa Cells , Humans , Intestines/microbiology , Molecular Sequence Data , Receptors, Cell Surface/genetics
18.
Microbiology (Reading) ; 153(Pt 6): 1743-1755, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17526832

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) is a major cause of infantile diarrhoea in developing countries. While colonizing the gut mucosa, EPEC triggers extensive actin-polymerization activity at the site of intimate bacterial attachment, which is mediated by avid interaction between the outer-membrane adhesin intimin and the type III secretion system (T3SS) effector Tir. The prevailing dogma is that actin polymerization by EPEC is achieved following tyrosine phosphorylation of Tir, recruitment of Nck and activation of neuronal Wiskott-Aldrich syndrome protein (N-WASP). In closely related enterohaemorrhagic E. coli (EHEC) O157 : H7, actin polymerization is triggered following recruitment of the T3SS effector TccP/EspF(U) (instead of Nck) and local activation of N-WASP. In addition to tccP, typical EHEC O157 : H7 harbour a pseudogene (tccP2). However, it has recently been found that atypical, sorbitol-fermenting EHEC O157 carries functional tccP and tccP2 alleles. Interestingly, intact tccP2 has been identified in the incomplete genome sequence of the prototype EPEC strain B171 (serotype O111 : H-), but it is missing from another prototype EPEC strain E2348/69 (O127 : H7). E2348/69 and B171 belong to two distinct evolutionary lineages of EPEC, termed EPEC 1 and EPEC 2, respectively. Here, it is reported that while both EPEC 1 and EPEC 2 triggered actin polymerization via the Nck pathway, tccP2 was found in 26 of 27 (96.2 %) strains belonging to EPEC 2, and in none of the 34 strains belonging to EPEC 1. It was shown that TccP2 was: (i) translocated by the locus of enterocyte effacement-encoded T3SS; (ii) localized at the tip of the EPEC 2-induced actin-rich pedestals in infected HeLa cells and human intestinal in vitro organ cultures ex vivo; and (iii) essential for actin polymerization in infected Nck-/- cells. Therefore, unlike strains belonging to EPEC 1, strains belonging to EPEC 2 can trigger actin polymerization using both Nck and TccP2 actin-polymerization signalling cascades.


Subject(s)
Actins/metabolism , Escherichia coli Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Escherichia coli Proteins/genetics , Gene Deletion , HeLa Cells , Humans , Intestine, Small/microbiology , Microscopy, Confocal , Microscopy, Electron, Scanning , Molecular Sequence Data , Oncogene Proteins/metabolism , Organ Culture Techniques , Polymerase Chain Reaction , Protein Transport , Sequence Alignment , Sequence Analysis, DNA
19.
Cell Microbiol ; 9(5): 1352-64, 2007 May.
Article in English | MEDLINE | ID: mdl-17474908

ABSTRACT

Tir, the translocated intimin receptor of enteropathogenic and enterohaemorrhagic Escherichia coli (EPEC and EHEC) and Citrobacter rodentium, is translocated into the host cell by a filamentous type III secretion system. Epithelial cell culture has demonstrated that Tir tyrosine phosphorylation is necessary for attaching effacing (A/E) lesion formation by EPEC and C. rodentium, but is not required by EHEC O157:H7. Recent in vivo work on C. rodentium has reported that Tir translocation, but not its phosphorylation, is necessary for colonization of the mouse colon. In this study we investigated the involvement of Tir and its tyrosine phosphorylation in EPEC and EHEC human intestinal colonization, N-WASP accumulation and F-actin recruitment using in vitro organ culture (IVOC). We showed that both EPEC and EHEC Tir are translocated into human intestinal epithelium during IVOC and that Tir is necessary for ex vivo intestinal colonization by both EPEC and EHEC. EPEC, but not EHEC, Tir is tyrosine phosphorylated but Tir phosphorylation-deficient mutants still colonize intestinal explants. While EPEC Tir recruits the host adaptor protein Nck to initiate N-WASP-Arp2/3-mediated actin polymerization, Tir derivatives deficient in tyrosine phosphorylation recruit N-WASP independently of Nck indicating the presence of a tyrosine phosphorylation-independent mechanism of A/E lesion formation and actin recruitment ex vivo by EPEC in man.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Intestinal Mucosa/microbiology , Oncogene Proteins/metabolism , Receptors, Cell Surface/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing , Cell Line, Tumor , Escherichia coli/growth & development , Escherichia coli/ultrastructure , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Humans , Immunoblotting , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Models, Biological , Organ Culture Techniques , Phosphorylation , Receptors, Cell Surface/genetics
20.
Appl Environ Microbiol ; 73(9): 3084-90, 2007 May.
Article in English | MEDLINE | ID: mdl-17351088

ABSTRACT

We used bovine intestinal organ culture to study infection by enterohemorrhagic Escherichia coli serogroups O157, O26, and O111. We show colonization and attaching and effacing lesion formation on explants derived from the ileum, colon, and rectum. Intimin and Tir were detected at the sites of adherent bacteria; Tir was essential for colonization.


Subject(s)
Bacterial Adhesion , Enterohemorrhagic Escherichia coli/physiology , Intestines/microbiology , Adhesins, Bacterial/metabolism , Animals , Cattle , Escherichia coli Proteins/metabolism , Immunohistochemistry , Intestinal Mucosa/metabolism , Receptors, Cell Surface/metabolism , Species Specificity , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...