Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 221
Filter
1.
Cell ; 140(4): 540-53, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20178746

ABSTRACT

Nascent transport intermediates detach from donor membranes by scission. This process can take place in the absence of dynamin, notably in clathrin-independent endocytosis, by mechanisms that are yet poorly defined. We show here that in cells scission of Shiga toxin-induced tubular endocytic membrane invaginations is preceded by cholesterol-dependent membrane reorganization and correlates with the formation of membrane domains on model membranes, suggesting that domain boundary forces are driving tubule membrane constriction. Actin triggers scission by inducing such membrane reorganization process. Tubule occurrence is indeed increased upon cellular depletion of the actin nucleator component Arp2, and the formation of a cortical actin shell in liposomes is sufficient to trigger the scission of Shiga toxin-induced tubules in a cholesterol-dependent but dynamin-independent manner. Our study suggests that membranes in tubular Shiga toxin-induced invaginations are poised to undergo actin-triggered reorganization leading to scission by a physical mechanism that may function independently from or in synergy with pinchase activity.


Subject(s)
Actins/metabolism , Cell Membrane/metabolism , Endocytosis , Cholesterol/metabolism , Dynamins/metabolism , HeLa Cells , Humans , Shiga Toxins/metabolism
2.
Nat Chem Biol ; 16(3): 327-336, 2020 03.
Article in English | MEDLINE | ID: mdl-32080624

ABSTRACT

The retrograde transport inhibitor Retro-2 has a protective effect on cells and in mice against Shiga-like toxins and ricin. Retro-2 causes toxin accumulation in early endosomes and relocalization of the Golgi SNARE protein syntaxin-5 to the endoplasmic reticulum. The molecular mechanisms by which this is achieved remain unknown. Here, we show that Retro-2 targets the endoplasmic reticulum exit site component Sec16A, affecting anterograde transport of syntaxin-5 from the endoplasmic reticulum to the Golgi. The formation of canonical SNARE complexes involving syntaxin-5 is not affected in Retro-2-treated cells. By contrast, the interaction of syntaxin-5 with a newly discovered binding partner, the retrograde trafficking chaperone GPP130, is abolished, and we show that GPP130 must indeed bind to syntaxin-5 to drive Shiga toxin transport from the endosomes to the Golgi. We therefore identify Sec16A as a druggable target and provide evidence for a non-SNARE function for syntaxin-5 in interaction with GPP130.


Subject(s)
Benzamides/metabolism , Qa-SNARE Proteins/metabolism , Thiophenes/metabolism , Vesicular Transport Proteins/metabolism , Benzamides/pharmacology , Biological Transport , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Golgi Apparatus/metabolism , HeLa Cells , Humans , Protein Transport , Ricin/metabolism , Shiga Toxin/metabolism , Shiga Toxins/metabolism , Thiophenes/pharmacology , Vesicular Transport Proteins/physiology
3.
PLoS Biol ; 16(11): e2006951, 2018 11.
Article in English | MEDLINE | ID: mdl-30481169

ABSTRACT

Glycosylation is a fundamental modification of proteins and membrane lipids. Toxins that utilize glycans as their receptors have served as powerful tools to identify key players in glycosylation processes. Here, we carried out Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9-mediated genome-wide loss-of-function screens using two related bacterial toxins, Shiga-like toxins (Stxs) 1 and 2, which use a specific glycolipid, globotriaosylceramide (Gb3), as receptors, and the plant toxin ricin, which recognizes a broad range of glycans. The Stxs screens identified major glycosyltransferases (GTs) and transporters involved in Gb3 biosynthesis, while the ricin screen identified GTs and transporters involved in N-linked protein glycosylation and fucosylation. The screens also identified lysosomal-associated protein transmembrane 4 alpha (LAPTM4A), a poorly characterized four-pass membrane protein, as a factor specifically required for Stxs. Mass spectrometry analysis of glycolipids and their precursors demonstrates that LAPTM4A knockout (KO) cells lack Gb3 biosynthesis. This requirement of LAPTM4A for Gb3 synthesis is not shared by its homolog lysosomal-associated protein transmembrane 4 beta (LAPTM4B), and switching the domains between them determined that the second luminal domain of LAPTM4A is required, potentially acting as a specific "activator" for the GT that synthesizes Gb3. These screens also revealed two Golgi proteins, Transmembrane protein 165 (TMEM165) and Transmembrane 9 superfamily member 2 (TM9SF2), as shared factors required for both Stxs and ricin. TMEM165 KO and TM9SF2 KO cells both showed a reduction in not only Gb3 but also other glycosphingolipids, suggesting that they are required for maintaining proper levels of glycosylation in general in the Golgi. In addition, TM9SF2 KO cells also showed defective endosomal trafficking. These studies reveal key Golgi proteins critical for regulating glycosylation and glycolipid synthesis and provide novel therapeutic targets for blocking Stxs and ricin toxicity.


Subject(s)
Ricin/genetics , Shiga Toxins/genetics , Bacterial Toxins/metabolism , CRISPR-Cas Systems , Endosomes/metabolism , Genome-Wide Association Study/methods , Glycolipids/metabolism , Glycosphingolipids , Glycosylation , Golgi Apparatus/metabolism , Golgi Apparatus/physiology , HEK293 Cells , HeLa Cells , Humans , Loss of Function Mutation/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/physiology , Oncogene Proteins/metabolism , Protein Transport , Ricin/metabolism , Shiga Toxins/metabolism , Trihexosylceramides/metabolism , Trihexosylceramides/physiology
4.
Glycobiology ; 30(3): 174-185, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31691795

ABSTRACT

Infections of the human intestinal tract with enterohemorrhagic Escherichia coli (EHEC) result in massive extraintestinal complications due to translocation of EHEC-released Shiga toxins (Stxs) from the gut into the circulation. Stx-mediated damage of the cerebral microvasculature raises serious brain dysfunction being the most frequent cause of acute mortality in patients suffering from severe EHEC infections. Stx2a and Stx2e are associated with heavy and mild course of infection, respectively. Stx2a preferentially binds to globotriaosylceramide (Gb3Cer, Galα1-4Galß1-4Glcß1-1Cer), while Stx2e prefers globotetraosylceramide (Gb4Cer, GalNAcß1-3Galα1-4Galß1-4Glcß1-1Cer). Both glycosphingolipids (GSLs) were detected in detergent-resistant membranes (DRMs) of primary human brain microvascular endothelial cells (pHBMECs) resembling microdomains of the plasma membrane. In this study, we show that Gb3Cer and Gb4Cer of pHBMECs with saturated C16:0, C22:0, and C24:0 fatty acids dominated in DRMs, corresponding to the liquid-ordered membrane phase, whereas lipoforms carrying unsaturated C24:1 and C24:2 fatty acids prevailed in the non-DRM fractions, which correspond to the liquid-disordered membrane phase. Similarly, a shift of the phospholipids from saturated lipoforms in the DRM to unsaturated species in the non-DRM fractions was observed. Real-time biomolecular interaction analysis using affinity-purified Stx2a and Stx2e, recorded with a surface acoustic wave (SAW) biosensor, evidenced high binding strength of both toxins toward DRMs and failure in interaction with non-DRMs. These results support the hypothesis of preferential binding of Stxs toward microdomains harboring GSL receptors carrying saturated fatty acids in their lipid anchors. Collectively, unraveling the precise mechanisms of Stx-microdomain interaction may help to develop antiadhesive compounds to combat Stx-mediated cellular injury.


Subject(s)
Brain/metabolism , Endothelial Cells/metabolism , Membrane Microdomains/metabolism , Shiga Toxins/metabolism , Endothelial Cells/chemistry , Humans , Membrane Microdomains/chemistry , Molecular Structure , Shiga Toxins/analysis , Time Factors
5.
Crit Rev Biotechnol ; 40(8): 1081-1097, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32811194

ABSTRACT

Shiga toxin-producing Escherichia coli (STEC) are usually found on food products due to contamination from the fecal origin, as their main environmental reservoir is considered to be the gut of ruminants. While this pathogen is far from the incidence of other well-known foodborne bacteria, the severity of STEC infections in humans has triggered global concerns as far as its incidence and control are concerned. Major control strategies for foodborne pathogens in food-related settings usually involve traditional sterilization/disinfection techniques. However, there is an increasing need for the development of further strategies to enhance the antimicrobial outcome, either on food-contact surfaces or directly in food matrices. Phages are considered to be a good alternative to control foodborne pathogens, with some phage-based products already cleared by the Food and Drug Administration (FDA) to be used in the food industry. In European countries, phage-based food decontaminants have already been used. Nevertheless, its broad use in the European Union is not yet possible due to the lack of specific guidelines for the approval of these products. Furthermore, some safety concerns remain to be addressed so that the regulatory requirements can be met. In this review, we present an overview of the main virulence factors of STEC and introduce phages as promising biocontrol agents for STEC control. We further present the regulatory constraints on the approval of phages for food applications and discuss safety concerns that are still impairing their use.


Subject(s)
Bacteriophages/physiology , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/virology , Animals , Europe , Feces/microbiology , Food Microbiology , Food Safety , Host Microbial Interactions/physiology , Humans , Life Cycle Stages , Shiga Toxins/genetics , Shiga-Toxigenic Escherichia coli/genetics , Virulence Factors
6.
J Cell Sci ; 128(15): 2891-902, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26071526

ABSTRACT

Endocytosis is an essential cellular process that is often hijacked by pathogens and pathogenic products. Endocytic processes can be classified into two broad categories, those that are dependent on clathrin and those that are not. The SNARE proteins VAMP2, VAMP3 and VAMP8 are internalized in a clathrin-dependent manner. However, the full scope of their endocytic behavior has not yet been elucidated. Here, we found that VAMP2, VAMP3 and VAMP8 are localized on plasma membrane invaginations and very early uptake structures that are induced by the bacterial Shiga toxin, which enters cells by clathrin-independent endocytosis. We show that toxin trafficking into cells and cell intoxication rely on these SNARE proteins. Of note, the cellular uptake of VAMP3 is increased in the presence of Shiga toxin, even when clathrin-dependent endocytosis is blocked. We therefore conclude that VAMP2, VAMP3 and VAMP8 are removed from the plasma membrane by non-clathrin-mediated pathways, in addition to by clathrin-dependent uptake. Moreover, our study identifies these SNARE proteins as the first transmembrane trafficking factors that functionally associate at the plasma membrane with the toxin-driven clathrin-independent invaginations during the uptake process.


Subject(s)
Endocytosis/physiology , Protein Transport/physiology , R-SNARE Proteins/metabolism , Shiga Toxin 1/pharmacology , Shiga Toxins/pharmacology , Vesicle-Associated Membrane Protein 2/metabolism , Vesicle-Associated Membrane Protein 3/metabolism , Cell Line , Cell Membrane/physiology , Clathrin/metabolism , ErbB Receptors/metabolism , HeLa Cells , Humans , Protein Binding/genetics , R-SNARE Proteins/genetics , RNA Interference , RNA, Small Interfering , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Shiga Toxins/metabolism , Transferrin/metabolism , Vesicle-Associated Membrane Protein 2/genetics , Vesicle-Associated Membrane Protein 3/genetics
7.
Environ Microbiol ; 19(5): 1836-1844, 2017 05.
Article in English | MEDLINE | ID: mdl-28127846

ABSTRACT

Shiga toxin producing Escherichia coli O157:H7 (STEC O157) is naturally found in the gastrointestinal tract of cattle and can cause severe disease in humans. There is limited understanding of the population dynamics and microevolution of STEC O157 at herd level. In this study, isolates from a closed beef herd of 23 cows were used to examine the population turnover in the herd. Of the nine STEC O157 clades previously described, clade 7 was found in 162 of the 169 isolates typed. Multiple locus variable number tandem repeat analysis (MLVA) differentiated 169 isolates into 33 unique MLVA types. Five predominant MLVA types were evident with most of the remaining types containing only a single isolate. MLVA data suggest that over time clonal replacement occurred within the herd. Genome sequencing of 18 selected isolates found that the isolates were divided into four lineages, representing four different 'clones' in the herd. Genome data confirmed clonal replacement over time and provided evidence of cross transmission of strains between cows. The findings enhanced our understanding of the population dynamics of STEC O157 in its natural host that will help developing effective control measures to prevent the spread of the pathogen to the human population.


Subject(s)
Escherichia coli O157/genetics , Escherichia coli O157/metabolism , Gastrointestinal Microbiome/genetics , Genome, Bacterial/genetics , Minisatellite Repeats/genetics , Molecular Typing/methods , Animals , Biological Evolution , Cattle , Escherichia coli Infections/epidemiology , Escherichia coli Infections/microbiology , Escherichia coli O157/isolation & purification , Female , Humans , Longitudinal Studies , Phylogeny , Polymorphism, Single Nucleotide/genetics , Population Dynamics , Red Meat/microbiology , Shiga Toxins/genetics , Shiga Toxins/metabolism
8.
Infect Immun ; 84(12): 3290-3301, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27600507

ABSTRACT

The A1 subunits of Shiga toxin 1 (Stx1A1) and Shiga toxin 2 (Stx2A1) interact with the conserved C termini of ribosomal-stalk P-proteins to remove a specific adenine from the sarcin/ricin loop. We previously showed that Stx2A1 has higher affinity for the ribosome and higher catalytic activity than Stx1A1. To determine if conserved arginines at the distal face of the active site contribute to the higher affinity of Stx2A1 for the ribosome, we mutated Arg172, Arg176, and Arg179 in both toxins. We show that Arg172 and Arg176 are more important than Arg179 for the depurination activity and toxicity of Stx1A1 and Stx2A1. Mutation of a single arginine reduced the depurination activity of Stx1A1 more than that of Stx2A1. In contrast, mutation of at least two arginines was necessary to reduce depurination by Stx2A1 to a level similar to that of Stx1A1. R176A and R172A/R176A mutations eliminated interaction of Stx1A1 and Stx2A1 with ribosomes and with the stalk, while mutation of Arg170 at the active site reduced the binding affinity of Stx1A1 and Stx2A1 for the ribosome, but not for the stalk. These results demonstrate that conserved arginines at the distal face of the active site are critical for interactions of Stx1A1 and Stx2A1 with the stalk, while a conserved arginine at the active site is critical for non-stalk-specific interactions with the ribosome. Arginine mutations at either site reduced ribosome interactions of Stx1A1 and Stx2A1 similarly, indicating that conserved arginines are critical for ribosome interactions but do not contribute to the higher affinity of Stx2A1 for the ribosome.


Subject(s)
Conserved Sequence , Escherichia coli Proteins/metabolism , Multienzyme Complexes/metabolism , Prephenate Dehydratase/metabolism , Ribosomes/metabolism , Saccharomyces/metabolism , Shiga Toxins/metabolism , Animals , Binding Sites , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial/physiology , Models, Molecular , Multienzyme Complexes/genetics , Mutation , Plasmids , Prephenate Dehydratase/genetics , Protein Binding , Protein Conformation , Protein Subunits , RNA, Fungal/metabolism , Rats , Ribosomes/chemistry , Saccharomyces/genetics , Shiga Toxins/chemistry
9.
Biotechnol Bioeng ; 113(8): 1639-46, 2016 08.
Article in English | MEDLINE | ID: mdl-26773973

ABSTRACT

The intracellular delivery of proteins with high efficiency in a receptor-specific manner is of great significance in molecular medicine and biotechnology, but remains a challenge. Herein, we present the development of a highly efficient and receptor-specific delivery platform for protein cargos by combining the receptor binding domain of Escherichia coli Shiga-like toxin and the translocation domain of Pseudomonas aeruginosa exotoxin A. We demonstrated the utility and efficiency of the delivery platform by showing a cytosolic delivery of diverse proteins both in vitro and in vivo in a receptor-specific manner. In particular, the delivery system was shown to be effective for targeting an intracellular protein and consequently suppressing the tumor growth in xenograft mice. The present platform can be widely used for intracellular delivery of diverse functional macromolecules with high efficiency in a receptor-specific manner. Biotechnol. Bioeng. 2016;113: 1639-1646. © 2016 Wiley Periodicals, Inc.


Subject(s)
ADP Ribose Transferases/metabolism , Bacterial Toxins/metabolism , Drug Delivery Systems/methods , Exotoxins/metabolism , Intracellular Space/metabolism , Protein Engineering/methods , Recombinant Fusion Proteins/metabolism , Shiga Toxins/metabolism , Virulence Factors/metabolism , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Cell Line, Tumor , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Exotoxins/chemistry , Exotoxins/genetics , Female , Mice , Mice, Inbred BALB C , Mice, Nude , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Shiga Toxins/chemistry , Shiga Toxins/genetics , Virulence Factors/chemistry , Virulence Factors/genetics , Pseudomonas aeruginosa Exotoxin A
10.
Appl Microbiol Biotechnol ; 100(4): 1597-1610, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26685676

ABSTRACT

Shiga toxins are a group of type 2 ribosome-inactivating proteins (RIPs) produced in several types of bacteria. The toxins possess an AB5 structure, which comprises a catalytic A chain with N-glycosidase activity, and five identical B chains and recognize and bind to the target cells with specific carbohydrate moieties. In humans, the major molecular target which recognizes the Shiga toxins is the Gb3 receptor, which is mainly expressed on the cell surface of endothelial cells of the intestine, kidney, and the brain. This causes these organs to be susceptible to the toxicity of Shiga toxins. When a person is infected by Shiga toxin-producing bacteria, the toxin is produced in the gut, translocated to the circulatory system, and carried to the target cells. Toxicity of the toxin causes inflammatory responses and severe cell damages in the intestine, kidneys, and brain, bringing about the hemolytic uremic syndrome (HUS), which can be fatal. The Shiga toxin requires a couple of steps to exert its toxicity to the target cells. After binding with the target cell surface receptor, the toxin requires a complicated process to be transported into the cytosol of the cell before it can approach the ribosomes. The mechanisms for the interactions of the toxin with the cells are described in this review. The consequences of the toxin on the cells are also discussed. It gives an overview of the steps for the toxin to be produced and transported, expression of catalytic activity, and the effects of the toxin on the target cells, as well as effects on the human body.


Subject(s)
Globosides/metabolism , Protein Synthesis Inhibitors/metabolism , Protein Synthesis Inhibitors/toxicity , Shiga Toxins/metabolism , Shiga Toxins/toxicity , Trihexosylceramides/metabolism , Brain/drug effects , Brain/pathology , Endothelial Cells/drug effects , Humans , Intestines/drug effects , Intestines/pathology , Kidney/drug effects , Kidney/pathology , Protein Synthesis Inhibitors/chemistry , Protein Transport , Ribosomes/drug effects , Shiga Toxins/chemistry
11.
Curr Microbiol ; 73(2): 265-72, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27155845

ABSTRACT

This work were aimed to (a) determination of minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of Carum copticum essential oil (EO) against Escherichia. coli O157:H7 in vitro Trypticase Soy Broth, (TSB) and in ground beef; (b) evaluation of the effect of sub-inhibitory concentrations (sub-MICs) of EO on the growth of bacterium in TSB over 72 h (at 35 °C) and ground beef over 9 days (at 4 °C); and (c) investigation of gene expression involved in Shiga toxins production using relative quantitative real-time PCR method. The MIC in broth and ground beef medium were determined as 0.05 (v/v) and 1.75 % (v/w), respectively. In comparison with control cultures, the EO concentration of 0.03 % in broth caused reduction of colony counting as 1.93, 1.79, and 2.62 log10 CFU ml(-1) after 24, 48, and 72 h at 35 °C, and similarly EO (0.75 %) in ground beef resulted to reduction of colony counting as 1.03, 0.92, 1.48, and 2.12 log10 CFU g (-1) after 2, 5, 7, and 9 days at 4 °C, respectively. An increase and decrease in gene expression were observed as result of EO addition (0.03 %) to broth and (0.5 %) to ground beef was noticed, respectively.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carum/chemistry , Escherichia coli O157/drug effects , Food Additives/pharmacology , Meat/microbiology , Oils, Volatile/pharmacology , Plant Oils/pharmacology , Shiga Toxins/genetics , Animals , Cattle , Colony Count, Microbial , Escherichia coli O157/genetics , Escherichia coli O157/metabolism , Shiga Toxins/metabolism
12.
New Microbiol ; 39(2): 119-23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27196550

ABSTRACT

The role of antibiotics in the treatment of Shiga-like toxin (Stx)-producing E. coli infection is still controversial. This study investigated the effects of colistin on Vero cell cytotoxicity caused by the enterohemorrhagic EC O157:H7, and the effects of colistin on Stx and endotoxin release by EC O157:H7. Vero cells were incubated with supernatant collected from EC O157:H7 cultured for 18 h without (control) or with various concentrations of colistin. In the absence of colistin, Vero cell viability after 48 h was 29.1±6.5%. Under the same conditions, the overnight presence of colistin reduced cytotoxicity to Vero cells (viability: 97±3.5 to 56.5±14.4% for colistin concentrations ≥MIC). Sub-MIC concentrations of colistin also provided partial protection (viability: 38.8±12.5 to 36.6±14% for 0.125 and 0.06 mcg/ml colistin, respectively). Endotoxins contributed to the cytotoxic effects on Vero cells since lower but still significant protection was observed when colistin was added directly to the supernatant collected from cultures of untreated EC O157:H7. Colistin reduced Stx release in a concentration-dependent manner, also at sub-MIC concentrations. Coincubation of the supernatant from EC O157:H7 cultures with colistin markedly reduced the endotoxin concentration at all doses investigated. In conclusion, colistin protects Vero cells from EC O157:H7 at supra- and sub-MIC concentrations by inhibiting Stx release and binding endotoxins. Colistin might be a valuable treatment for clinically severe forms of EC O157:H7 infection.


Subject(s)
Cell Survival/drug effects , Colistin/pharmacology , Endotoxins/chemistry , Escherichia coli O157/drug effects , Shiga Toxins/metabolism , Animals , Chlorocebus aethiops , Colistin/chemistry , Escherichia coli O157/metabolism , Shiga Toxins/genetics , Vero Cells
13.
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
14.
Microbiology (Reading) ; 161(Pt 3): 451-62, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25479836

ABSTRACT

Shiga toxins are the main virulence factors of a group of Escherichia coli strains [Shiga toxin-producing E. coli (STEC)] that cause severe human diseases, such as haemorrhagic colitis and haemolytic-uraemic syndrome. The Shiga toxin family comprises several toxin subtypes, which have been differentially related to clinical manifestations. In addition, the phages that carry the Shiga toxin genes (stx phages) are also diverse. These phages play an important role not only in the dissemination of Shiga toxin genes and the emergence of new STEC strains, but also in the regulation of Shiga toxin production. Consequently, differences in stx phages may affect the dissemination of stx genes as well as the virulence of STEC strains. In addition to presenting an overview of Shiga toxins and stx phages, in this review we highlight current knowledge about the diversity of stx phages, with emphasis on its impact on STEC virulence. We consider that this diversity should be taken into account when developing STEC infection treatments and diagnostic approaches, and when conducting STEC control in reservoirs.


Subject(s)
Bacteriophages/metabolism , Escherichia coli Infections/microbiology , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Shiga-Toxigenic Escherichia coli/virology , Animals , Bacteriophages/genetics , Humans , Shiga Toxins/toxicity , Shiga-Toxigenic Escherichia coli/genetics , Shiga-Toxigenic Escherichia coli/pathogenicity , Virulence
15.
Traffic ; 13(8): 1140-59, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22540229

ABSTRACT

Retrograde trafficking transports proteins, lipids and toxins from the plasma membrane to the Golgi and endoplasmic reticulum (ER). To reach the Golgi, these cargos must transit the endosomal system, consisting of early endosomes (EE), recycling endosomes, late endosomes and lysosomes. All cargos pass through EE, but may take different routes to the Golgi. Retromer-dependent cargos bypass the late endosomes to reach the Golgi. We compared how two very different retromer-dependent cargos negotiate the endosomal sorting system. Shiga toxin B, bound to the external layer of the plasma membrane, and chimeric CD8-mannose-6-phosphate receptor (CI-M6PR), which is anchored via a transmembrane domain. Both appear to pass through the recycling endosome. Ablation of the recycling endosome diverted both of these cargos to an aberrant compartment and prevented them from reaching the Golgi. Once in the recycling endosome, Shiga toxin required EHD1 to traffic to the TGN, while the CI-M6PR was not significantly dependent on EHD1. Knockdown of retromer components left cargo in the EE, suggesting that it is required for retrograde exit from this compartment. This work establishes the recycling endosome as a required step in retrograde traffic of at least these two retromer-dependent cargos. Along this pathway, retromer is associated with EE to recycling endosome traffic, while EHD1 is associated with recycling endosome to TGN traffic of STxB.


Subject(s)
Endosomes/metabolism , Golgi Apparatus/metabolism , Protein Transport/physiology , Vesicular Transport Proteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , Microscopy, Fluorescence , RNA Interference , Receptor, IGF Type 2 , Receptors, Cytoplasmic and Nuclear/metabolism , Shiga Toxins/metabolism , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/genetics , trans-Golgi Network/metabolism
16.
Am J Pathol ; 182(4): 1227-38, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23402998

ABSTRACT

Enterohemorrhagic Escherichia coli cause approximately 1.5 million infections globally with 176,000 cases occurring in the United States annually from ingesting contaminated food, most frequently E. coli O157:H7 in ground beef or fresh produce. In severe cases, the painful prodromal hemorrhagic colitis is complicated by potentially lethal hemolytic uremic syndrome (HUS), particularly in children. Bacterial Shiga-like toxins (Stx1, Stx2) are primarily responsible for HUS and the kidney and neurologic damage that ensue. Small animal models are hampered by the inability to reproduce HUS with thrombotic microangiopathy, hemolytic anemia, and acute kidney injury. Earlier, we showed that nonhuman primates (Papio) recapitulated clinical HUS after Stx challenge and that novel therapeutic intervention rescued the animals. Here, we present detailed light and electron microscopic pathology examination of the kidneys from these Stx studies. Stx1 challenge resulted in more severe glomerular endothelial injury, whereas the glomerular injury after Stx2 also included prominent mesangiolysis and an eosinophilic inflammatory infiltration. Both toxins induced glomerular platelet-rich thrombi, interstitial hemorrhage, and tubular injury. Analysis of kidney and other organs for inflammation biomarkers showed a striking chemotactic profile, with extremely high mRNA levels for IL-8, monocyte chemoattractant protein 1, and macrophage inflammatory protein 1α and elevated urine chemokines at 48 hours after challenge. These observations give unique insight into the pathologic consequences of each toxin in a near human setting and present potential pathways for therapeutic intervention.


Subject(s)
Chemotaxis , Enterohemorrhagic Escherichia coli/physiology , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/pathology , Kidney/pathology , Papio/microbiology , Shiga Toxins/metabolism , Animals , Chemokines/genetics , Chemokines/urine , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Endothelial Cells/pathology , Endothelial Cells/ultrastructure , Eosinophils/pathology , Gene Expression Regulation , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/urine , Humans , Inflammation/pathology , Kidney/metabolism , Kidney/microbiology , Kidney/ultrastructure , Mesangial Cells/metabolism , Mesangial Cells/microbiology , Mesangial Cells/pathology , Mesangial Cells/ultrastructure , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Shiga Toxin 1/metabolism , Shiga Toxin 2/metabolism
17.
Foodborne Pathog Dis ; 11(3): 248-55, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24313785

ABSTRACT

Different structures related to biofilm formation by Shiga toxin-producing Escherichia coli (STEC), particularly O157 strains, have been described, but there are few data regarding their involvement in non-O157 strains. The aim of this study was to determine the ability of 14 O157 and 8 non-O157 strains isolated from bovine hide and carcass to interact with biotic and abiotic surfaces and also to evaluate the role of different adhesins. Biofilm formation assays showed that four O157 and two non-O157 strains were able to adhere to glass, and that only one O157 strain adhered to polystyrene. Reverse transcriptase-polymerase chain reaction was carried out using biofilm-forming strains to determine the expression of antigen 43 (Ag43), curli, type 1 fimbriae, STEC autotransporter contributing to biofilm formation (Sab), calcium-binding antigen 43 homologue (Cah), and autotransporter protein of enterohemorrhagic E. coli (EhaA). Most of these structures were expressed under biofilm conditions. However, the lack of Ag43 in one non-O157 strain, as well as Cah and EhaA in two O157 strains, suggests that other adhesins are involved in biofilm formation in these strains. Despite the fact that adherence to HeLa cells was detected in 20 strains (91%), it was not possible to correlate biofilm formation with adherence patterns. Invasiveness in T84 and Caco-2 cells was observed in four and three O157 strains, respectively. Altogether, we showed that there are different sets of genes involved in the interactions of STEC with biotic and abiotic surfaces. Interestingly, one O157 strain that was able to form biofilm on both glass and polystyrene also adhered to and invaded human cells, indicating an important route for its persistence in the environment and interaction with the host. Additionally, the ability of non-O157 strains not carrying the LEE pathogenicity island to form biofilm highlights an industrial and health problem that cannot be neglected.


Subject(s)
Biofilms/growth & development , Escherichia coli Proteins/genetics , Shiga-Toxigenic Escherichia coli/physiology , Adhesins, Bacterial/genetics , Animals , Bacterial Adhesion/physiology , Cattle , Cell Line, Tumor , Escherichia coli O157/isolation & purification , Escherichia coli O157/physiology , Fimbriae, Bacterial/genetics , Glass , Humans , Meat/microbiology , Polystyrenes , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/isolation & purification , Skin/microbiology
18.
Foodborne Pathog Dis ; 11(6): 439-46, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24796582

ABSTRACT

Shiga toxin-producing Escherichia coli (STEC) are foodborne pathogens of worldwide importance, but a shortage of data exists for STEC isolation from India. Therefore, an epidemiological and environmental study that covers a large geographic area in north India was conducted. Ruminant stool samples (n=650) were collected from 59 dairies. Meat samples (n=450) were collected from local abattoirs and the main slaughterhouse of the region. Additionally, 600 human cases of diarrhea and hemolytic uremic syndrome were screened for STEC. Isolates were characterized for the virulence gene profiles and for the serogroups and were submitted to molecular typing by the multilocus variable-number tandem-repeat analysis (MLVA). Overall, 12.3% of animal stool samples and 6.3% of mutton samples (n=160) were positive for STEC. Additionally, STEC were isolated from 1.7% and 1.6% of watery (n=290) and bloody (n=310) stool specimens, respectively. Animal stool isolates were significantly more prevalent in hilly areas (p<0.05) than in plain areas. Polymerase chain reaction demonstrated the presence of stx1, stx2, hly, espP, saa, toxB, and iha genes in 117 (83.5%), 94 (67.1%), 77 (55%), 33 (23%), 62 (44.2%), 29 (20.7%), and 51 (36%) of the isolates, respectively. Five new serogroups (O55, O33, O173, O165, and O136) are being reported for the first time from India. Four isolates from serogroup O103 were found in mutton and stool specimens of cattle and humans (n=160). One isolate from serogroup O104 was isolated from a mutton sample. MLVA suggested the potential transmission of STEC from contaminated meat and bovine sources. This study confirms the frequent contamination of mutton samples (24%), whereas chicken and pork samples were negative for STEC. This study demonstrates the presence of STEC that carry a large repertoire of virulence genes and the potential transmission of STEC from contaminated mutton and animal stools in north India.


Subject(s)
Escherichia coli Infections/microbiology , Foodborne Diseases/microbiology , Meat/microbiology , Sheep, Domestic/microbiology , Shiga Toxins/analysis , Shiga-Toxigenic Escherichia coli/isolation & purification , Virulence Factors/analysis , Abattoirs , Animals , Cattle , Child, Preschool , Dairying , Diarrhea/etiology , Escherichia coli Infections/epidemiology , Escherichia coli Infections/physiopathology , Feces/chemistry , Feces/microbiology , Female , Food Contamination , Foodborne Diseases/epidemiology , Foodborne Diseases/physiopathology , Hemolytic-Uremic Syndrome/etiology , Humans , India/epidemiology , Infant , Male , Meat/analysis , Molecular Typing , Prevalence , Shiga Toxins/genetics , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/classification , Shiga-Toxigenic Escherichia coli/growth & development , Shiga-Toxigenic Escherichia coli/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
19.
Sci Rep ; 14(1): 6043, 2024 03 13.
Article in English | MEDLINE | ID: mdl-38472311

ABSTRACT

Shiga toxins (Stx) produced by pathogenic bacteria can cause mild to severe diseases in humans. Thus, the analysis of such toxins is of utmost importance. As an AB5 toxin, Stx consist of a catalytic A-subunit acting as a ribosome-inactivating protein (RIP) and a B-pentamer binding domain. In this study we synthesized the subunits and holotoxins from Stx and Stx2a using different cell-free systems, namely an E. coli- and CHO-based cell-free protein synthesis (CFPS) system. The functional activity of the protein toxins was analyzed in two ways. First, activity of the A-subunits was assessed using an in vitro protein inhibition assay. StxA produced in an E. coli cell-free system showed significant RIP activity at concentrations of 0.02 nM, whereas toxins synthesized in a CHO cell-free system revealed significant activity at concentrations of 0.2 nM. Cell-free synthesized StxA2a was compared to StxA2a expressed in E. coli cells. Cell-based StxA2a had to be added at concentrations of 20 to 200 nM to yield a significant RIP activity. Furthermore, holotoxin analysis on cultured HeLa cells using an O-propargyl-puromycin assay showed significant protein translation reduction at concentrations of 10 nM and 5 nM for cell-free synthesized toxins derived from E. coli and CHO systems, respectively. Overall, these results show that Stx can be synthesized using different cell-free systems while remaining functionally active. In addition, we were able to use CFPS to assess the activity of different Stx variants which can further be used for RIPs in general.


Subject(s)
Escherichia coli , Shiga Toxins , Humans , Shiga Toxins/metabolism , Escherichia coli/genetics , Cell-Free System/metabolism , HeLa Cells , Protein Biosynthesis
20.
Infect Immun ; 81(8): 2753-60, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23690406

ABSTRACT

The two major forms of Shiga toxin, Stx1 and Stx2, use the glycolipid globotriaosylceramide (Gb3) as their cellular receptor. Stx1 primarily recognizes the Pk-trisaccharide portion and has three Pk binding sites per B monomer. The Stx2a subtype requires glycolipid residues in addition to Pk. We synthesized analogs of Pk to examine the binding preferences of Stx1 and Stx2 subtypes a to d. Furthermore, to determine how many binding sites must be engaged, the Pk analogues were conjugated to biotinylated mono- and biantennary platforms, allowing for the display of two to four Pk analogues per streptavidin molecule. Stx binding to Pk analogues immobilized on streptavidin-coated plates was assessed by enzyme-linked immunosorbent assay (ELISA). Stx1, but not the Stx2 subtypes, bound to native Pk. Stx2a and Stx2c bound to the Pk analog with a terminal GalNAc (NAc-Pk), while Stx1, Stx2b, and Stx2d did not bind to this analog. Interestingly, the purified Stx2d B subunit bound to NAc-Pk, suggesting that the A subunit of Stx2d interferes with binding. Disaccharide analogs (Galα1-4Gal, GalNAcα1-4Gal, and Galα1-4GalNAc) did not support the binding of any of the Stx forms, indicating that the trisaccharide is necessary for binding. Studies with monoantennary and biantennary analogs and mixtures suggest that Stx1, Stx2a, and Stx2c need to engage at least three Pk analogues for effective binding. To our knowledge, this is the first study examining the minimum number of Pk analogs required for effective binding and the first report documenting the role of the A subunit in influencing Stx2 binding.


Subject(s)
Shiga Toxins/chemistry , Trihexosylceramides/chemistry , Trisaccharides/chemistry , Amino Acid Sequence , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Molecular Sequence Data , Protein Binding , Shiga Toxins/metabolism , Trisaccharides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL