Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Toxins (Basel) ; 13(10)2021 09 28.
Article in English | MEDLINE | ID: mdl-34678982

ABSTRACT

Shiga toxins (Stxs), also known as Shiga-like toxins (SLT) or verotoxins (VT), constitute a family of structurally and functionally related cytotoxic proteins produced by the enteric pathogens Shigella dysenteriae type 1 and Stx-producing Escherichia coli (STEC). Infection with these bacteria causes bloody diarrhea and other pathological manifestations that can lead to HUS (hemolytic and uremic syndrome). At the cellular level, Stxs bind to the cellular receptor Gb3 and inhibit protein synthesis by removing an adenine from the 28S rRNA. This triggers multiple cellular signaling pathways, including the ribotoxic stress response (RSR), unfolded protein response (UPR), autophagy and apoptosis. Stxs cause several pathologies of major public health concern, but their specific targeting of host cells and efficient delivery to the cytosol could potentially be exploited for biomedical purposes. Moreover, high levels of expression have been reported for the Stxs receptor, Gb3/CD77, in Burkitt's lymphoma (BL) cells and on various types of solid tumors. These properties have led to many attempts to develop Stxs as tools for biomedical applications, such as cancer treatment or imaging, and several engineered Stxs are currently being tested. We provide here an overview of these studies.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Shiga Toxins/pharmacology , Apoptosis , Autophagy , Drug Delivery Systems , Humans , Ribosomes/drug effects , Shiga Toxins/chemistry , Signal Transduction/drug effects , Trihexosylceramides/metabolism
2.
Toxins (Basel) ; 13(3)2021 03 18.
Article in English | MEDLINE | ID: mdl-33803852

ABSTRACT

Shiga toxins (Stxs) are classic bacterial toxins and major virulence factors of toxigenic Shigella dysenteriae and enterohemorrhagic Escherichia coli (EHEC). These toxins recognize a glycosphingolipid globotriaosylceramide (Gb3/CD77) as their receptor and inhibit protein synthesis in cells by cleaving 28S ribosomal RNA. They are the major cause of life-threatening complications such as hemolytic uremic syndrome (HUS), associated with severe cases of EHEC infection, which is the leading cause of acute kidney injury in children. The threat of Stxs is exacerbated by the lack of toxin inhibitors and effective treatment for HUS. Here, we briefly summarize the Stx structure, subtypes, in vitro and in vivo models, Gb3 expression and HUS, and then introduce recent studies using CRISPR-Cas9-mediated genome-wide screens to identify the host cell factors required for Stx action. We also summarize the latest progress in utilizing and engineering Stx components for biomedical applications.


Subject(s)
Escherichia coli Infections/metabolism , Hemolytic-Uremic Syndrome/metabolism , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Trihexosylceramides/metabolism , Animals , CRISPR-Cas Systems , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/microbiology , Host-Pathogen Interactions , Humans , Immunotoxins/therapeutic use , Models, Molecular , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Protein Conformation , Shiga Toxins/chemistry , Shiga Toxins/genetics , Shiga Toxins/therapeutic use , Shiga-Toxigenic Escherichia coli/genetics , Structure-Activity Relationship
3.
Methods Mol Biol ; 2132: 463-474, 2020.
Article in English | MEDLINE | ID: mdl-32306353

ABSTRACT

Shiga toxin (Stx) is a major virulence factor of enterohemorrhagic Escherichia coli (E. coli). Stx consists of one enzymatic A subunit and five B subunits (StxB) that are involved in binding. The StxB pentamer specifically recognizes a glycosphingolipid, globotriaosylceramide (Gb3), as a receptor; therefore, it can be used as a probe to detect Gb3. This chapter describes the preparation of recombinant Stx1B proteins using E. coli, their conjugation with fluorescent dyes, and their application for flow cytometry. The prepared fluorescent StxB proteins bound to cells of several lines, including the HeLa human cervix adenocarcinoma cell line and the THP-1 human monocytic leukemia cell line. Furthermore, the probe was useful for confirmation of several sphingolipid-deficient HeLa cell lines that were constructed using genome editing.


Subject(s)
Enterohemorrhagic Escherichia coli/metabolism , Rhodamines/chemistry , Shiga Toxins/isolation & purification , Sulfonic Acids/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/isolation & purification , Flow Cytometry , Gene Editing , HeLa Cells , Humans , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Shiga Toxins/chemistry , Sphingolipids/deficiency , THP-1 Cells
4.
Food Sci Technol Int ; 25(6): 491-496, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30862194

ABSTRACT

Antimicrobial treatments could help to decrease the transmission of microorganisms to beef carcasses and abattoir environments. The aim of this study was to evaluate the effectiveness of interventions in reducing Shiga toxin genes (stx1 and stx2) presence in a commercial abattoir. Intervention measures included the application of electrolytically generated hypochlorous acid to steer pens (experiment 1), chlorinated water, electrolytically generated hypochlorous acid, and isoclor to steer pens (experiment 2), electrolytically generated hypochlorous acid to knocking pens (experiment 3), and aqueous ozone and electrolytically generated hypochlorous acid onto beef carcasses (experiment 4). Detection of stx in samples was performed with BAX® System Real-Time PCR Assay. Our results showed that treatment with pressurized electrolytically generated hypochlorous acid and isoclor were effective to reduce stx presence from hides on steer pens. Although there is no single strategy to ensure the reduction of stx presence in a commercial abattoir, the combined application of several antimicrobial interventions would be ideal.


Subject(s)
Abattoirs , Chlorine Compounds/pharmacology , Electrochemical Techniques , Housing, Animal , Shiga Toxins/chemistry , Animals , Cattle , Decontamination/methods , Disinfection , Feces/microbiology , Pressure
5.
Chemphyschem ; 18(13): 1822-1830, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28608625

ABSTRACT

We develop an excitable automata model of a protein verotoxin and demonstrate that logic gates and circuits are realised in the model via interacting patterns of excitation. By sampling potential input pairs of nodes, we calculate frequencies of logic gates which occurred in the verotoxin model for various parameters of node excitation rules. We show that overall the gates can be arranged in the following hierarchy of descending frequencies: AND>OR>AND-NOT>XOR. We demonstrate realisations of one-bit half-adder and controlled-not gates and estimate memory capacity of the verotoxin molecule.


Subject(s)
Computers, Molecular , Shiga Toxins/chemistry , Models, Molecular
6.
Toxins (Basel) ; 9(2)2017 02 03.
Article in English | MEDLINE | ID: mdl-28165371

ABSTRACT

Shiga toxins consist of an A-moiety and five B-moieties able to bind the neutral glycosphingolipid globotriaosylceramide (Gb3) on the cell surface. To intoxicate cells efficiently, the toxin A-moiety has to be cleaved by furin and transported retrogradely to the Golgi apparatus and to the endoplasmic reticulum. The enzymatically active part of the A-moiety is then translocated to the cytosol, where it inhibits protein synthesis and in some cell types induces apoptosis. Protection of cells can be provided either by inhibiting binding of the toxin to cells or by interfering with any of the subsequent steps required for its toxic effect. In this article we provide a brief overview of the interaction of Shiga toxins with cells, describe some compounds and conditions found to protect cells against Shiga toxins, and discuss whether they might also provide protection in animals and humans.


Subject(s)
Antidotes/pharmacology , Bacterial Proteins/antagonists & inhibitors , Dysentery, Bacillary/prevention & control , Hemolytic-Uremic Syndrome/prevention & control , Shiga Toxins/antagonists & inhibitors , Shiga-Toxigenic Escherichia coli/drug effects , Shigella dysenteriae/drug effects , Animals , Apoptosis , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/microbiology , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/microbiology , Host-Pathogen Interactions , Humans , Protein Biosynthesis , Protein Conformation , Protein Transport , Shiga Toxins/chemistry , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity , Shigella dysenteriae/metabolism , Shigella dysenteriae/pathogenicity , Structure-Activity Relationship , Trihexosylceramides/metabolism
7.
Nat Nanotechnol ; 11(12): 1112-1119, 2016 12.
Article in English | MEDLINE | ID: mdl-27548358

ABSTRACT

Functionalization of quantum dots (QDs) with a single biomolecular tag using traditional approaches in bulk solution has met with limited success. DNA polyhedra consist of an internal void bounded by a well-defined three-dimensional structured surface. The void can house cargo and the surface can be functionalized with stoichiometric and spatial precision. Here, we show that monofunctionalized QDs can be realized by encapsulating QDs inside DNA icosahedra and functionalizing the DNA shell with an endocytic ligand. We deployed the DNA-encapsulated QDs for real-time imaging of three different endocytic ligands-folic acid, galectin-3 (Gal3) and the Shiga toxin B-subunit (STxB). Single-particle tracking of Gal3- or STxB-functionalized QD-loaded DNA icosahedra allows us to monitor compartmental dynamics along endocytic pathways. These DNA-encapsulated QDs, which bear a unique stoichiometry of endocytic ligands, represent a new class of molecular probes for quantitative imaging of endocytic receptor dynamics.


Subject(s)
DNA/chemistry , Endocytosis/physiology , Molecular Imaging/methods , Quantum Dots/chemistry , Animals , Cricetulus , Dynamic Light Scattering , Endosomes/metabolism , Fibroblasts/metabolism , Folic Acid/chemistry , Galectin 3/analysis , Galectin 3/chemistry , Galectin 3/metabolism , Mice , Microscopy, Electron, Transmission , Molecular Imaging/instrumentation , Shiga Toxins/analysis , Shiga Toxins/chemistry , Shiga Toxins/metabolism
8.
Drug Des Devel Ther ; 10: 3363-3378, 2016.
Article in English | MEDLINE | ID: mdl-27789937

ABSTRACT

The aim of this study was to investigate the bacteriostatic and bactericidal effects of diminazene aceturate (DA) against five strains of pathogenic bacteria and two strains of nonpathogenic bacteria. The results showed that 5 µg/mL of DA suppressed the growth of pathogenic Escherichia coli by as much as 77% compared with the controls. Enterohemorrhagic E. coli EDL933 (an E. coli O157:H7 strain) was the most sensitive to DA with a minimum inhibitory concentration of 20 µg/mL. Additional investigations showed that DA induced the highest level of intracellular reactive oxygen species in EDL933. A positive correlation between the reactive oxygen species levels and DA concentration was demonstrated. DA (5 µg/mL) was also a potent uncoupler, inducing a stationary phase collapse (70%-75%) in both strains of E. coli O157:H7. Further investigation showed that the collapse was due to the NaCl:DA ratio in the broth and was potassium ion dependent. A protease screening assay was conducted to elucidate the underlying mechanism. It was found that at neutral pH, the hydrolysis of H-Asp-pNA increased by a factor of 2-3 in the presence of DA, implying that DA causes dysregulation of the proton motive force and a decrease in cellular pH. Finally, a commercial verotoxin test showed that DA did not significantly increase toxin production in EDL933 and was a suitable antibacterial agent for Shiga-toxin-producing E. coli.


Subject(s)
Anti-Bacterial Agents/pharmacology , Diminazene/analogs & derivatives , Escherichia coli O157/drug effects , Peptide Hydrolases/chemistry , Shiga Toxin/antagonists & inhibitors , Shiga Toxins/adverse effects , Anti-Bacterial Agents/chemistry , Diminazene/chemistry , Diminazene/pharmacology , Escherichia coli O157/chemistry , High-Throughput Screening Assays , Shiga Toxins/chemistry
9.
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
10.
Toxins (Basel) ; 8(3)2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26999205

ABSTRACT

Shiga toxins (Stxs) produced by Shiga toxin-producing bacteria Shigella dysenteriae serotype 1 and select serotypes of Escherichia coli are primary virulence factors in the pathogenesis of hemorrhagic colitis progressing to potentially fatal systemic complications, such as hemolytic uremic syndrome and central nervous system abnormalities. Current therapeutic options to treat patients infected with toxin-producing bacteria are limited. The structures of Stxs, toxin-receptor binding, intracellular transport and the mode of action of the toxins have been well defined. However, in the last decade, numerous studies have demonstrated that in addition to being potent protein synthesis inhibitors, Stxs are also multifunctional proteins capable of activating multiple cell stress signaling pathways, which may result in apoptosis, autophagy or activation of the innate immune response. Here, we briefly present the current understanding of Stx-activated signaling pathways and provide a concise review of therapeutic applications to target tumors by engineering the toxins.


Subject(s)
Shiga Toxins , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Protein Conformation , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , Protein Synthesis Inhibitors/therapeutic use , Shiga Toxins/chemistry , Shiga Toxins/pharmacology , Shiga Toxins/therapeutic use , Signal Transduction/drug effects , Stress, Physiological
11.
Rapid Commun Mass Spectrom ; 30(6): 671-80, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-26864518

ABSTRACT

RATIONAL: Analysis of bacteria by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOFMS) often relies upon sample preparation methods that result in cell lysis, e.g. bead-beating. However, Shiga toxin-producing Escherichia coli (STEC) can undergo bacteriophage-induced cell lysis triggered by antibiotic exposure that may allow greater selectivity of the proteins extracted. METHODS: We have developed a sample preparation method for selective extraction of bacteriophage-encoded proteins and specifically Shiga toxins 1 and 2 (Stx1 & 2) expressed from STEC strains induced by DNA-damaging antibiotics. STEC strains were cultured overnight on agar supplemented with ciprofloxacin, mitomycin-C or an iron chelator to induce the bacteriophage lytic cycle with concomitant expression and release of Stx1 and/or Stx2. Sample preparation relied exclusively on bacteriophage lysis for release Stx into the extraction solution. RESULTS: Three clinical STEC strains were analyzed by matrix-assisted laser desorption/ionization tandem time-of-flight mass spectrometry (MALDI-TOF-TOF-MS/MS) and top-down proteomics analysis: E. coli O157:H7 strain EDL933, E. coli O91:H21 strain B2F1 and E. coli O26:H11 strain ECRC #05.2217. The B-subunit of Stx1a of EDL933 was detected and identified even though it was ~100-fold less abundant than the B-subunit of Stx2a that had been identified previously for this strain. Two bacteriophage-encoded proteins were also identified: L0117 and L0136. The B-subunits of Stx2d of strain B2F1 and Stx1a of strain ECRC #05.2217 were also detected and identified. CONCLUSIONS: Bacteriophage lysis appeared to enhance the detection sensitivity of Stx for these STEC strains compared to previous work using mechanical lysis. Detection/identification of other bacteriophage-encoded proteins (beyond Stx) tends to support the hypothesis of Stx release by bacteriophage cell lysis.


Subject(s)
Proteomics/methods , Shiga Toxins/analysis , Shiga Toxins/chemistry , Shiga-Toxigenic Escherichia coli/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Bacteriophages , Molecular Sequence Data , Shiga-Toxigenic Escherichia coli/virology
12.
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
13.
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
14.
Anal Chem ; 86(11): 5541-6, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24797274

ABSTRACT

Small compounds cannot bind simultaneously to two antibodies, and thus, their immunodetection is limited to competitive formats in which the analyte is indirectly quantitated by measuring the unoccupied antibody binding sites using a competing reporter. This limitation can be circumvented by using phage-borne peptides selected for their ability to specifically react with the analyte-antibody immunocomplex, which allows the detection of these small molecules in a noncompetitive format (PHAIA) with increased sensitivity and a positive readout. In an effort to find substitutes for the phage particles in PHAIA, we explore the use of the B subunit of the Shiga-like toxin of Escherichia coli, also known as verotoxin (VTX), as a scaffold for multivalent display of anti-immunocomplex peptides. Using the herbicides molinate and clomazone as model compounds, we built peptide-VTX recombinant chimeras that were produced in the periplasmic space of E. coli as soluble pentamers, as confirmed by multiangle light scattering analysis. These multivalent constructs, which we termed nanopeptamers, were conjugated to a tracer enzyme and used to detect the herbicide-antibody complex in an ELISA format. The VTX-nanopeptamer assays performed with over a 10-fold increased sensitivity and excellent recovery from spiked surface and mineral water samples. The carbon black-labeled peptide-VTX nanopeptamers showed great potential for the development of a lateral-flow test for small molecules with a visual positive readout that allowed the detection of up to 2.5 ng/mL of clomazone.


Subject(s)
Peptides/chemistry , Shiga Toxin/chemistry , Shiga-Toxigenic Escherichia coli/chemistry , Azepines/analysis , Enzyme-Linked Immunosorbent Assay , Herbicides/analysis , Immunotoxins/chemistry , Isoxazoles/analysis , Mutagenesis , Oxazolidinones/analysis , Protein Conformation , Shiga Toxins/chemistry , Thiocarbamates/analysis , Viral Fusion Proteins/chemistry , Water Pollutants, Chemical/analysis
15.
Cell Mol Life Sci ; 71(21): 4285-300, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24740796

ABSTRACT

Shiga toxin-producing Escherichia coli bacteria cause hemorrhagic colitis and hemolytic uremic syndrome in humans. Currently, only supportive treatment is available for diagnosed patients. We show here that 24-h pretreatment with an ether lipid precursor, the alkylglycerol sn-1-O-hexadecylglycerol (HG), protects HEp-2 cells against Shiga toxin and Shiga toxin 2. Also the endothelial cell lines HMEC-1 and HBMEC are protected against Shiga toxins after HG pretreatment. In contrast, the corresponding acylglycerol, DL-α-palmitin, has no effect on Shiga toxicity. Although HG treatment provides a strong protection (~30 times higher IC50) against Shiga toxin, only a moderate reduction in toxin binding was observed, suggesting that retrograde transport of the toxin from the plasma membrane to the cytosol is perturbed. Furthermore, endocytosis of Shiga toxin and retrograde sorting from endosomes to the Golgi apparatus remain intact, but transport from the Golgi to the endoplasmic reticulum is inhibited by HG treatment. As previously described, HG reduces the total level of all quantified glycosphingolipids to 50-70% of control, including the Shiga toxin receptor globotriaosylceramide (Gb3), in HEp-2 cells. In accordance with this, we find that interfering with Gb3 biosynthesis by siRNA-mediated knockdown of Gb3 synthase for 24 h causes a similar cytotoxic protection and only a moderate reduction in toxin binding (to 70% of control cells). Alkylglycerols, including HG, have been administered to humans for investigation of therapeutic roles in disorders where ether lipid biosynthesis is deficient, as well as in cancer therapy. Further studies may reveal if HG can also have a therapeutic potential in Shiga toxin-producing E. coli infections.


Subject(s)
Glycerol/chemistry , Glyceryl Ethers/chemistry , Shiga Toxins/chemistry , Biological Transport , Biotinylation , Cell Line , Cell Membrane/metabolism , Cytosol/metabolism , Endocytosis , Endoplasmic Reticulum/metabolism , Escherichia coli/metabolism , Ether/chemistry , Glycosphingolipids/chemistry , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Inhibitory Concentration 50 , Lipids/chemistry , Palmitic Acid/chemistry , Shiga Toxin/chemistry , Trihexosylceramides/chemistry
16.
Nihon Saikingaku Zasshi ; 68(3): 299-311, 2013.
Article in Japanese | MEDLINE | ID: mdl-23985936

ABSTRACT

Bacterial AB5 toxins are proteins, produced by pathogenic bacteria including of Vibrio cholerae, Shigella dysenteriae, and enterohaemorrhagic Escherichia coli, which are usually released into the extracellular medium and cause disease by killing or altering the metabolism of target eukaryotic cells. The toxins are usually composed of one A subunit (a toxic domain) and five B subunits (a receptor-binding domain). This article overviews the characteristics and mode of actions of AB5 toxins including cholera toxin, Shiga-like toxin, and subtilase cytotoxin, and highlights current topics related to the roles of the effectors in promoting bacterial infection.


Subject(s)
Cholera Toxin/toxicity , Enterohemorrhagic Escherichia coli/metabolism , Escherichia coli Proteins/toxicity , Shiga Toxins/toxicity , Subtilisins/toxicity , Vibrio cholerae/metabolism , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cholera Toxin/antagonists & inhibitors , Cholera Toxin/biosynthesis , Cholera Toxin/chemistry , Disease Outbreaks , Endoplasmic Reticulum Stress/drug effects , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/chemistry , Foodborne Diseases/epidemiology , Foodborne Diseases/microbiology , Humans , Macrophages/immunology , Phagocytosis/drug effects , Protein Structure, Tertiary , Shiga Toxins/antagonists & inhibitors , Shiga Toxins/biosynthesis , Shiga Toxins/chemistry , Subtilisins/antagonists & inhibitors , Subtilisins/biosynthesis , Subtilisins/chemistry , Vaccines, Attenuated , Vibrio cholerae/pathogenicity
17.
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
18.
Bioorg Med Chem Lett ; 22(24): 7448-50, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23142616

ABSTRACT

A functional detection assay was developed for Escherichia coli secreted shiga-like toxin based on antibody capture and visualization with a multivalent galabiose ligand. It was possible to detect verotoxin in medically relevant E. coli samples in a dose dependent fashion. This method is a new step towards measuring functional protein levels in complex mixtures, which can be used for diagnostic purposes in a clinical setting.


Subject(s)
Antibodies/chemistry , Antigens, Bacterial/analysis , Antigens, Bacterial/chemistry , Disaccharides/chemistry , Shiga Toxins/analysis , Shiga Toxins/chemistry , Antibodies/immunology , Antigen-Antibody Reactions , Antigens, Bacterial/immunology , Binding Sites , Biotin/chemistry , Escherichia coli/chemistry , Ligands , Serum Albumin, Bovine/chemistry , Shiga Toxins/immunology
19.
Toxicon ; 60(6): 1085-107, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22960449

ABSTRACT

Shiga toxins are virulence factors produced by the bacteria Shigella dysenteriae and certain strains of Escherichia coli. There is currently no available treatment for disease caused by these toxin-producing bacteria, and understanding the biology of the Shiga toxins might be instrumental in addressing this issue. In target cells, the toxins efficiently inhibit protein synthesis by inactivating ribosomes, and they may induce signaling leading to apoptosis. To reach their cytoplasmic target, Shiga toxins are endocytosed and transported by a retrograde pathway to the endoplasmic reticulum, before the enzymatically active moiety is translocated to the cytosol. The toxins thereby serve as powerful tools to investigate mechanisms of intracellular transport. Although Shiga toxins are a serious threat to human health, the toxins may be exploited for medical purposes such as cancer therapy or imaging.


Subject(s)
Shiga Toxins/chemistry , Virulence Factors , Apoptosis , Biological Transport , Endocytosis , Endoplasmic Reticulum/metabolism , Escherichia coli/chemistry , Shiga Toxins/genetics , Shiga Toxins/isolation & purification , Shigella dysenteriae/chemistry , Signal Transduction
20.
Blood ; 120(5): 1143-9, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22718838

ABSTRACT

Shiga toxin (Stx) causes diarrhea-associated hemolytic uremic syndrome by damaging renal microvascular endothelium. The pentameric B subunits of Stx types 1 and 2 (Stx1B and Stx2B) are sufficient to stimulate acute VWF secretion from endothelial cells, but Stx1B and Stx2B exert distinct effects on Ca(2+) and cAMP pathways. Therefore, we investigated other signaling components in StxB-induced VWF exocytosis. Incubation of HUVECs with StxB transiently increased phospholipase D (PLD) activity. Inhibition of PLD activity or shRNA-mediated PLD1 knockdown abolished StxB-induced VWF secretion. In addition, treatment with StxB triggered actin polymerization, enhanced endothelial monolayer permeability, and activated RhoA. PLD activation and VWF secretion induced by Stx1B were abolished on protein kinase Cα (PKCα) inhibition or gene silencing but were only moderately reduced by Rho or Rho kinase inhibitors. Conversely, PLD activation and VWF exocytosis induced by Stx2B were reduced by Rho/Rho kinase inhibitors and dominant-negative RhoA, whereas attenuation of PKCα did not affect either process. Another PLD1 activator, ADP-ribosylation factor 6, was involved in VWF secretion induced by Stx1B or Stx2B, but not histamine. These data indicate that Stx1B and Stx2B induce acute VWF secretion in a PLD1-dependent manner but do so by differentially modulating PKCα, RhoA, and ADP-ribosylation factor 6.


Subject(s)
Phospholipase D/physiology , Shiga Toxins/pharmacology , von Willebrand Factor/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Cells, Cultured , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Enzyme Activation/physiology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Permeability/drug effects , Phospholipase D/antagonists & inhibitors , Phospholipase D/genetics , Phospholipase D/metabolism , Protein Kinase C-alpha/metabolism , Protein Subunits/metabolism , Protein Subunits/pharmacology , RNA, Small Interfering/pharmacology , Shiga Toxin 2/pharmacology , Shiga Toxins/chemistry , Time Factors , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...