Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Front Microbiol ; 14: 1198945, 2023.
Article in English | MEDLINE | ID: mdl-37303786

ABSTRACT

Outer membrane vesicles (OMVs) carrying virulence factors of enterohemorrhagic Escherichia coli (EHEC) are assumed to play a role in the pathogenesis of life-threatening hemolytic uremic syndrome (HUS). However, it is unknown if and how OMVs, which are produced in the intestinal lumen, cross the intestinal epithelial barrier (IEB) to reach the renal glomerular endothelium, the major target in HUS. We investigated the ability of EHEC O157 OMVs to translocate across the IEB using a model of polarized Caco-2 cells grown on Transwell inserts and characterized important aspects of this process. Using unlabeled or fluorescently labeled OMVs, tests of the intestinal barrier integrity, inhibitors of endocytosis, cell viability assay, and microscopic techniques, we demonstrated that EHEC O157 OMVs translocated across the IEB. OMV translocation involved both paracellular and transcellular pathways and was significantly increased under simulated inflammatory conditions. In addition, translocation was not dependent on OMV-associated virulence factors and did not affect viability of intestinal epithelial cells. Importantly, translocation of EHEC O157 OMVs was confirmed in human colonoids thereby supporting physiological relevance of OMVs in the pathogenesis of HUS.

2.
Dis Model Mech ; 15(4)2022 04 01.
Article in English | MEDLINE | ID: mdl-35302159

ABSTRACT

The gut microbiota plays a crucial role in protecting against enteric infection. However, the underlying mechanisms are largely unknown owing to a lack of suitable experimental models. Although most gut commensals are anaerobic, intestinal epithelial cells require oxygen for survival. In addition, most intestinal cell lines do not produce mucus, which provides a habitat for the microbiota. Here, we have developed a microaerobic, mucus-producing vertical diffusion chamber (VDC) model and determined the influence of Limosilactobacillus reuteri and Ruminococcus gnavus on enteropathogenic Escherichia coli (EPEC) infection. Optimization of the culture medium enabled bacterial growth in the presence of mucus-producing T84/LS174T cells. Whereas L. reuteri diminished EPEC growth and adhesion to T84/LS174T and mucus-deficient T84 epithelia, R. gnavus only demonstrated a protective effect in the presence of LS174T cells. Reduced EPEC adherence was not associated with altered type III secretion pore formation. In addition, co-culture with L. reuteri and R. gnavus dampened EPEC-induced interleukin 8 secretion. The microaerobic mucin-producing VDC system will facilitate investigations into the mechanisms underpinning colonization resistance and aid the development of microbiota-based anti-infection strategies. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Anaerobiosis , Epithelial Cells/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Humans , Intestinal Mucosa/metabolism , Intestines/microbiology
3.
Methods Mol Biol ; 2291: 273-283, 2021.
Article in English | MEDLINE | ID: mdl-33704758

ABSTRACT

The environment in the human intestine is low in oxygen. This affects virulence gene expression of enteropathogens including Shiga toxin-producing E. coli (STEC) and enables mucosal colonization by oxygen-sensitive commensal bacteria. To simulate the oxygen-restricted milieu at the intestinal epithelium, we have developed a vertical diffusion chamber model (VDC) which allows infection of polarized human intestinal epithelia under microaerobic conditions. In this chapter, we present a detailed protocol for performing STEC infections in the VDC system and subsequent analysis of STEC pathogenesis.


Subject(s)
Intestinal Mucosa , Models, Biological , Oxygen/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology
4.
Appl Environ Microbiol ; 86(24)2020 11 24.
Article in English | MEDLINE | ID: mdl-33008822

ABSTRACT

The third E. coli and the Mucosal Immune System (ECMIS) meeting was held at Ghent University in Belgium from 2 to 5 June 2019. It brought together an international group of scientists interested in mechanisms of colonization, host response, and vaccine development. ECMIS distinguishes itself from related meetings on these enteropathogens by providing a greater emphasis on animal health and disease and covering a broad range of pathotypes, including enterohemorrhagic, enteropathogenic, enterotoxigenic, enteroaggregative, and extraintestinal pathogenic Escherichia coli As it is well established that the genus Shigella represents a subspecies of E. coli, these organisms along with related enteroinvasive E. coli are also included. In addition, Tannerella forsythia, a periodontal pathogen, was presented as an example of a pathogen which uses its surface glycans for mucosal interaction. This review summarizes several highlights from the 2019 meeting and major advances to our understanding of the biology of these pathogens and their impact on the host.


Subject(s)
Escherichia coli Infections/immunology , Escherichia coli/physiology , Immunity, Mucosal , Gram-Negative Bacterial Infections/immunology , Tannerella forsythia/physiology
5.
Sci Rep ; 10(1): 7475, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32366874

ABSTRACT

Enteroaggregative E. coli (EAEC) are a major cause of diarrhoea worldwide. Due to their heterogeneity and carriage in healthy individuals, identification of diagnostic virulence markers for pathogenic strains has been difficult. In this study, we have determined phenotypic and genotypic differences between EAEC strains of sequence types (STs) epidemiologically associated with asymptomatic carriage (ST31) and diarrhoeal disease (ST40). ST40 strains demonstrated significantly enhanced intestinal adherence, biofilm formation, and pro-inflammatory interleukin-8 secretion compared with ST31 isolates. This was independent of whether strains were derived from diarrhoea patients or healthy controls. Whole genome sequencing revealed differences in putative virulence genes encoding aggregative adherence fimbriae, E. coli common pilus, flagellin and EAEC heat-stable enterotoxin 1. Our results indicate that ST40 strains have a higher intrinsic potential of human pathogenesis due to a specific combination of virulence-related factors which promote host cell colonization and inflammation. These findings may contribute to the development of genotypic and/or phenotypic markers for EAEC strains of high virulence.


Subject(s)
Biofilms/growth & development , Escherichia coli Infections , Escherichia coli Proteins , Escherichia coli , Virulence Factors , Escherichia coli/pathogenicity , Escherichia coli/physiology , Escherichia coli Infections/genetics , Escherichia coli Infections/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , HeLa Cells , Humans , Virulence Factors/genetics , Virulence Factors/metabolism
6.
PLoS Pathog ; 15(8): e1008031, 2019 08.
Article in English | MEDLINE | ID: mdl-31465434

ABSTRACT

Enterohemorrhagic E. coli (EHEC) is a human intestinal pathogen that causes hemorrhagic colitis and hemolytic uremic syndrome. No vaccines or specific therapies are currently available to prevent or treat these infections. EHEC tightly attaches to the intestinal epithelium by injecting the intimin receptor Tir into the host cell via a type III secretion system (T3SS). In this project, we identified a camelid single domain antibody (nanobody), named TD4, that recognizes a conserved Tir epitope overlapping the binding site of its natural ligand intimin with high affinity and stability. We show that TD4 inhibits attachment of EHEC to cultured human HeLa cells by preventing Tir clustering by intimin, activation of downstream actin polymerization and pedestal formation. Furthermore, we demonstrate that TD4 significantly reduces EHEC adherence to human colonic mucosa in in vitro organ cultures. Altogether, these results suggest that nanobody-based therapies hold potential in the development of much needed treatment and prevention strategies against EHEC infection.


Subject(s)
Bacterial Adhesion/physiology , Colon/metabolism , Enterohemorrhagic Escherichia coli/physiology , Escherichia coli Infections/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Single-Domain Antibodies/pharmacology , Amino Acid Sequence , Animals , Bacterial Adhesion/drug effects , Binding Sites , Camelus , Colon/microbiology , Colon/pathology , Enterohemorrhagic Escherichia coli/drug effects , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Escherichia coli Proteins/immunology , Escherichia coli Proteins/metabolism , HeLa Cells , Humans , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Sequence Homology , Single-Domain Antibodies/immunology
7.
FEMS Microbiol Rev ; 43(5): 457-489, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31162610

ABSTRACT

A close symbiotic relationship exists between the intestinal microbiota and its host. A critical component of gut homeostasis is the presence of a mucus layer covering the gastrointestinal tract. Mucus is a viscoelastic gel at the interface between the luminal content and the host tissue that provides a habitat to the gut microbiota and protects the intestinal epithelium. The review starts by setting up the biological context underpinning the need for experimental models to study gut bacteria-mucus interactions in the digestive environment. We provide an overview of the structure and function of intestinal mucus and mucins, their interactions with intestinal bacteria (including commensal, probiotics and pathogenic microorganisms) and their role in modulating health and disease states. We then describe the characteristics and potentials of experimental models currently available to study the mechanisms underpinning the interaction of mucus with gut microbes, including in vitro, ex vivo and in vivo models. We then discuss the limitations and challenges facing this field of research.


Subject(s)
Gastrointestinal Microbiome , Gastrointestinal Tract/physiology , Microbial Interactions , Mucus/microbiology , Animals , Gastrointestinal Tract/microbiology , Homeostasis , Host Microbial Interactions , Humans , In Vitro Techniques , Intestinal Mucosa/microbiology , Mice , Models, Animal , Mucins/chemistry , Mucins/metabolism , Rats
8.
Front Microbiol ; 10: 879, 2019.
Article in English | MEDLINE | ID: mdl-31114553

ABSTRACT

Interactions of anaerobic gut bacteria, such as Clostridium difficile, with the intestinal mucosa have been poorly studied due to challenges in culturing anaerobes with the oxygen-requiring gut epithelium. Although gut colonization by C. difficile is a key determinant of disease outcome, precise mechanisms of mucosal attachment and spread remain unclear. Here, using human gut epithelial monolayers co-cultured within dual environment chambers, we demonstrate that C. difficile adhesion to gut epithelial cells is accompanied by a gradual increase in bacterial numbers. Prolonged infection causes redistribution of actin and loss of epithelial integrity, accompanied by production of C. difficile spores, toxins, and bacterial filaments. This system was used to examine C. difficile interactions with the commensal Bacteroides dorei, and interestingly, C. difficile growth is significantly reduced in the presence of B. dorei. Subsequently, we have developed novel models containing a myofibroblast layer, in addition to the epithelium, grown on polycarbonate or three-dimensional (3D) electrospun scaffolds. In these more complex models, C. difficile adheres more efficiently to epithelial cells, as compared to the single epithelial monolayers, leading to a quicker destruction of the epithelium. Our study describes new controlled environment human gut models that enable host-anaerobe and pathogen-commensal interaction studies in vitro.

9.
Cell Microbiol ; 21(6): e13012, 2019 06.
Article in English | MEDLINE | ID: mdl-30673154

ABSTRACT

Enteroaggregative Escherichia coli (EAEC) are important intestinal pathogens causing acute and persistent diarrhoeal illness worldwide. Although many putative EAEC virulence factors have been identified, their association with pathogenesis remains unclear. As environmental cues can modulate bacterial virulence, we investigated the effect of oxygen and human intestinal epithelium on EAEC virulence gene expression to determine the involvement of respective gene products in intestinal colonisation and pathogenesis. Using in vitro organ culture of human intestinal biopsies, we established the colonic epithelium as the major colonisation site of EAEC strains 042 and 17-2. We subsequently optimised a vertical diffusion chamber system with polarised T84 colon carcinoma cells for EAEC infection and showed that oxygen induced expression of the global regulator AggR, aggregative adherence fimbriae, E. coli common pilus, EAST-1 toxin, and dispersin in EAEC strain 042 but not in 17-2. Furthermore, the presence of T84 epithelia stimulated additional expression of the mucinase Pic and the toxins HlyE and Pet. This induction was dependent on physical host cell contact and did not require AggR. Overall, these findings suggest that EAEC virulence in the human gut is modulated by environmental signals including oxygen and the intestinal epithelium.


Subject(s)
Colon/microbiology , Escherichia coli Infections/microbiology , Escherichia coli/pathogenicity , Intestinal Mucosa/microbiology , Oxygen/metabolism , Virulence Factors/metabolism , Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/metabolism , Bacterial Toxins/metabolism , Cell Line, Tumor , Colon/ultrastructure , Enterotoxins/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Gene Expression Regulation, Bacterial , Hemolysin Proteins/metabolism , Host-Pathogen Interactions , Humans , Intestinal Mucosa/ultrastructure , Intestine, Small/microbiology , Polysaccharide-Lyases/metabolism , Serine Endopeptidases/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Virulence Factors/genetics
10.
Microorganisms ; 6(4)2018 Nov 19.
Article in English | MEDLINE | ID: mdl-30463258

ABSTRACT

Enterohaemorrhagic Escherichia coli (EHEC) is a major foodborne pathogen responsible for human diseases ranging from diarrhoea to life-threatening complications. Survival of the pathogen and modulation of virulence gene expression along the human gastrointestinal tract (GIT) are key features in bacterial pathogenesis, but remain poorly described, due to a paucity of relevant model systems. This review will provide an overview of the in vitro and in vivo studies investigating the effect of abiotic (e.g., gastric acid, bile, low oxygen concentration or fluid shear) and biotic (e.g., gut microbiota, short chain fatty acids or host hormones) parameters of the human gut on EHEC survival and/or virulence (especially in relation with motility, adhesion and toxin production). Despite their relevance, these studies display important limitations considering the complexity of the human digestive environment. These include the evaluation of only one single digestive parameter at a time, lack of dynamic flux and compartmentalization, and the absence of a complex human gut microbiota. In a last part of the review, we will discuss how dynamic multi-compartmental in vitro models of the human gut represent a novel platform for elucidating spatial and temporal modulation of EHEC survival and virulence along the GIT, and provide new insights into EHEC pathogenesis.

11.
Microbiology (Reading) ; 164(4): 509-516, 2018 04.
Article in English | MEDLINE | ID: mdl-29533744

ABSTRACT

Shiga toxin-producing Escherichia coli (STEC) are characterized by the release of potent Shiga toxins (Stx), which are associated with severe intestinal and renal disease. Although all STEC strains produce Stx, only a few serotypes cause infection in humans. To determine which virulence traits in vitro are linked to human disease in vivo, 13 Stx2a-producing STEC strains of seropathotype (SPT) A or B (associated with severe human intestinal disease and outbreaks) and 6 strains of SPT D or E (rarely or not linked to human disease) were evaluated in a microaerobic human colonic epithelial infection model. All SPT strains demonstrated similar growth, colonization of polarized T84 colon carcinoma cells and Stx release into the medium. In contrast, Stx translocation across the T84 cell monolayer was significantly lower in SPT group DE compared to SPT group AB strains. Further experiments showed that Stx penetration occurred via a transcellular pathway and was independent of bacterial type III secretion and attaching and effacing lesion formation. These results suggest that the extent of Stx transcytosis across the gut epithelium may represent an important indicator of STEC pathogenicity for humans.


Subject(s)
Escherichia coli Infections/microbiology , Intestinal Mucosa/metabolism , Shiga Toxin 2/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity , Transcytosis , Virulence Factors/metabolism , Anaerobiosis , Animals , Cell Line, Tumor , Chlorocebus aethiops , Escherichia coli Infections/metabolism , Humans , Intestinal Mucosa/microbiology , Serogroup , Shiga-Toxigenic Escherichia coli/growth & development , Shiga-Toxigenic Escherichia coli/isolation & purification , Vero Cells , Virulence
12.
J Infect Dis ; 218(6): 979-990, 2018 08 14.
Article in English | MEDLINE | ID: mdl-29471349

ABSTRACT

Background: Epidemiological studies point to the gut as a key reservoir of multidrug resistant Escherichia coli multilocus sequence type 131 (ST131), a globally dominant pathogenic clone causing urinary tract and bloodstream infections. Here we report a detailed investigation of its intestinal lifestyle. Methods: Clinical ST131 isolates and type 1 fimbriae null mutants were assessed for colonization of human intestinal epithelia and in mouse intestinal colonization models. Mouse gut tissue underwent histologic analysis for pathology and ST131 localization. Key findings were corroborated in mucus-producing human cell lines and intestinal biopsy specimens. Results: ST131 strains adhered to and invaded human intestinal epithelial cells more than probiotic and commensal strains. The reference ST131 strain EC958 established persistent intestinal colonization in mice, and expression of type 1 fimbriae mediated higher colonization levels. Bacterial loads were highest in the distal parts of the mouse intestine and did not cause any obvious pathology. Further analysis revealed that EC958 could bind to both mucus and underlying human intestinal epithelia. Conclusions: ST131 strains can efficiently colonize the mammalian gut and persist long term. Type 1 fimbriae enhance ST131 intestinal colonization, suggesting that mannosides, currently developed as therapeutics for bladder infections and Crohn's disease, could also be used to limit intestinal ST131 reservoirs.


Subject(s)
Drug Resistance, Multiple, Bacterial , Escherichia coli Infections/metabolism , Escherichia coli/pathogenicity , Intestines/microbiology , Animals , Bacterial Adhesion , Bacterial Load , Caco-2 Cells , Cell Line , Epithelial Cells/cytology , Epithelial Cells/microbiology , Escherichia coli/classification , Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Female , Fimbriae, Bacterial/metabolism , Humans , Intestines/cytology , Mice
13.
PLoS Pathog ; 13(10): e1006706, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29084270

ABSTRACT

Enteropathogenic E. coli (EPEC) is a human pathogen that causes acute and chronic pediatric diarrhea. The hallmark of EPEC infection is the formation of attaching and effacing (A/E) lesions in the intestinal epithelium. Formation of A/E lesions is mediated by genes located on the pathogenicity island locus of enterocyte effacement (LEE), which encode the adhesin intimin, a type III secretion system (T3SS) and six effectors, including the essential translocated intimin receptor (Tir). Seventeen additional effectors are encoded by genes located outside the LEE, in insertion elements and prophages. Here, using a stepwise approach, we generated an EPEC mutant lacking the entire effector genes (EPEC0) and intermediate mutants. We show that EPEC0 contains a functional T3SS. An EPEC mutant expressing intimin but lacking all the LEE effectors but Tir (EPEC1) was able to trigger robust actin polymerization in HeLa cells and mucin-producing intestinal LS174T cells. However, EPEC1 was unable to form A/E lesions on human intestinal in vitro organ cultures (IVOC). Screening the intermediate mutants for genes involved in A/E lesion formation on IVOC revealed that strains lacking non-LEE effector/s have a marginal ability to form A/E lesions. Furthermore, we found that Efa1/LifA proteins are important for A/E lesion formation efficiency in EPEC strains lacking multiple effectors. Taken together, these results demonstrate the intricate relationships between T3SS effectors and the essential role non-LEE effectors play in A/E lesion formation on mucosal surfaces.


Subject(s)
Adhesins, Bacterial/metabolism , Enterocytes/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/genetics , Intestinal Mucosa/microbiology , Adhesins, Bacterial/genetics , Carrier Proteins/metabolism , Genomic Islands , Humans , Intestinal Mucosa/metabolism
14.
Cell Microbiol ; 19(6)2017 06.
Article in English | MEDLINE | ID: mdl-28054754

ABSTRACT

Enterohaemorrhagic Escherichia coli (EHEC) is a major foodborne pathogen and tightly adheres to human colonic epithelium by forming attaching/effacing lesions. To reach the epithelial surface, EHEC must penetrate the thick mucus layer protecting the colonic epithelium. In this study, we investigated how EHEC interacts with the intestinal mucus layer using mucin-producing LS174T colon carcinoma cells and human colonic mucosal biopsies. The level of EHEC binding and attaching/effacing lesion formation in LS174T cells was higher compared to mucin-deficient colon carcinoma cell lines, and initial adherence was independent of the presence of flagellin, Escherichia coli common pilus, or long polar fimbriae. Although EHEC infection did not affect gene expression of secreted mucins, it resulted in reduced MUC2 glycoprotein levels. This effect was dependent on the catalytic activity of the secreted metalloprotease StcE, which reduced the inner mucus layer and thereby promoted EHEC access and binding to the epithelium in vitro and ex vivo. Given the lack of efficient therapies against EHEC infection, StcE may represent a suitable target for future treatment and prevention strategies.


Subject(s)
Bacterial Adhesion/physiology , Escherichia coli Infections/pathology , Escherichia coli O157/pathogenicity , Escherichia coli Proteins/metabolism , Intestinal Mucosa/microbiology , Metalloendopeptidases/metabolism , Mucus/metabolism , Bacterial Adhesion/genetics , Caco-2 Cells , Cell Line , Colon/microbiology , Colon/pathology , Escherichia coli Infections/microbiology , Escherichia coli O157/genetics , Escherichia coli O157/metabolism , Escherichia coli Proteins/genetics , Fimbriae, Bacterial/metabolism , Flagellin/metabolism , HT29 Cells , Humans , Intestinal Mucosa/pathology , Metalloendopeptidases/genetics , Mucin-2/metabolism
15.
Article in English | MEDLINE | ID: mdl-27446815

ABSTRACT

Enterohemorrhagic E.coli (EHEC) is an important foodborne pathogen in the developed world and can cause life-threatening disease particularly in children. EHEC persists in the human gut by adhering intimately to colonic epithelium and forming characteristic attaching/effacing lesions. In this study, we investigated the innate immune response to EHEC infection with particular focus on antimicrobial peptide and protein expression by colonic epithelium. Using a novel human colonic biopsy model and polarized T84 colon carcinoma cells, we found that EHEC infection induced expression of human ß-defensin 2 (hBD2), whereas hBD1, hBD3, LL-37, and lysozyme remained unchanged. Infection with specific EHEC deletion mutants demonstrated that this was dependent on flagellin, and apical exposure to purified flagellin was sufficient to stimulate hBD2 and also interleukin (IL)-8 expression ex vivo and in vitro. Flagellin-mediated hBD2 induction was significantly reduced by inhibitors of NF-κB, MAP kinase p38 and JNK but not ERK1/2. Interestingly, IL-8 secretion by polarized T84 cells was vectorial depending on the side of stimulation, and apical exposure to EHEC or flagellin resulted in apical IL-8 release. Our results demonstrate that EHEC only induces a modest immune response in human colonic epithelium characterized by flagellin-dependent induction of hBD2 and low levels of IL-8.


Subject(s)
Colon/metabolism , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Escherichia coli Proteins/metabolism , Flagellin/pharmacology , beta-Defensins/drug effects , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Anti-Infective Agents/pharmacology , Bacterial Adhesion , Biopsy , Cell Line, Tumor , Colon/microbiology , Colonic Neoplasms , Escherichia coli Proteins/genetics , Flagellin/genetics , Gene Expression Regulation, Bacterial , Humans , Immunity, Innate , Interleukin-8/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , NF-kappa B/metabolism , Sequence Deletion
16.
Front Microbiol ; 7: 244, 2016.
Article in English | MEDLINE | ID: mdl-26973622

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) is a major cause of diarrheal infant death in developing countries, and probiotic bacteria have been shown to provide health benefits in gastrointestinal infections. In this study, we have investigated the influence of the gut symbiont Lactobacillus reuteri on EPEC adherence to the human intestinal epithelium. Different host cell model systems including non-mucus-producing HT-29 and mucus-producing LS174T intestinal epithelial cell lines as well as human small intestinal biopsies were used. Adherence of L. reuteri to HT-29 cells was strain-specific, and the mucus-binding proteins CmbA and MUB increased binding to both HT-29 and LS174T cells. L. reuteri ATCC PTA 6475 and ATCC 53608 significantly inhibited EPEC binding to HT-29 but not LS174T cells. While pre-incubation of LS174T cells with ATCC PTA 6475 did not affect EPEC attaching/effacing (A/E) lesion formation, it increased the size of EPEC microcolonies. ATCC PTA 6475 and ATCC 53608 binding to the mucus layer resulted in decreased EPEC adherence to small intestinal biopsy epithelium. Our findings show that L. reuteri reduction of EPEC adhesion is strain-specific and has the potential to target either the epithelium or the mucus layer, providing further rationale for the selection of probiotic strains.

17.
Infect Immun ; 83(3): 942-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25534942

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) is an important foodborne pathogen causing gastroenteritis and more severe complications, such as hemorrhagic colitis and hemolytic uremic syndrome. Pathology is most pronounced in the colon, but to date there is no direct clinical evidence showing EHEC binding to the colonic epithelium in patients. In this study, we investigated EHEC adherence to the human colon by using in vitro organ culture (IVOC) of colonic biopsy samples and polarized T84 colon carcinoma cells. We show for the first time that EHEC colonizes human colonic biopsy samples by forming typical attaching and effacing (A/E) lesions which are dependent on EHEC type III secretion (T3S) and binding of the outer membrane protein intimin to the translocated intimin receptor (Tir). A/E lesion formation was dependent on oxygen levels and suppressed under oxygen-rich culture conditions routinely used for IVOC. In contrast, EHEC adherence to polarized T84 cells occurred independently of T3S and intimin and did not involve Tir translocation into the host cell membrane. Colonization of neither biopsy samples nor T84 cells was significantly affected by expression of Shiga toxins. Our study suggests that EHEC colonizes and forms stable A/E lesions on the human colon, which are likely to contribute to intestinal pathology during infection. Furthermore, care needs to be taken when using cell culture models, as they might not reflect the in vivo situation.


Subject(s)
Adhesins, Bacterial/genetics , Escherichia coli O157/genetics , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Intestinal Mucosa/microbiology , Receptors, Cell Surface/genetics , Shiga Toxins/genetics , Adhesins, Bacterial/metabolism , Bacterial Adhesion , Cell Line, Tumor , Colon/microbiology , Colon/pathology , Escherichia coli O157/drug effects , Escherichia coli O157/metabolism , Escherichia coli O157/pathogenicity , Escherichia coli Proteins/metabolism , Humans , Intestinal Mucosa/pathology , Oxygen/pharmacology , Protein Binding , Receptors, Cell Surface/metabolism , Shiga Toxins/metabolism
18.
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
19.
Toxins (Basel) ; 3(6): 626-39, 2011 06.
Article in English | MEDLINE | ID: mdl-22069729

ABSTRACT

After ingestion via contaminated food or water, enterohaemorrhagic E. coli colonises the intestinal mucosa and produces Shiga toxins (Stx). No Stx-specific secretion system has been described so far, and it is assumed that Stx are released into the gut lumen after bacterial lysis. Human intestinal epithelium does not express the Stx receptor Gb3 or other Stx binding sites, and it remains unknown how Stx cross the intestinal epithelial barrier and gain access to the systemic circulation. This review summarises current knowledge about the influence of the intestinal environment on Stx production and release, Stx interaction with intestinal epithelial cells and intracellular uptake, and toxin translocation into underlying tissues. Furthermore, it highlights gaps in understanding that need to be addressed by future research.


Subject(s)
Enterohemorrhagic Escherichia coli , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Intestinal Mucosa/microbiology , Shiga Toxin 1/toxicity , Shiga Toxin 2/toxicity , Animals , Bacterial Outer Membrane Proteins/metabolism , Bacteriophages , Binding Sites , Enterohemorrhagic Escherichia coli/pathogenicity , Enterohemorrhagic Escherichia coli/virology , Humans , Intestinal Mucosa/drug effects , Protein Transport , Shiga Toxin 1/genetics , Shiga Toxin 1/metabolism , Shiga Toxin 2/genetics , Shiga Toxin 2/metabolism , Trihexosylceramides/genetics
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...