Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Gut Microbes ; 14(1): 2122667, 2022.
Article in English | MEDLINE | ID: mdl-36138514

ABSTRACT

Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli (EHEC) cause gastrointestinal infection and, in severe cases, hemolytic uremic syndrome which may lead to death. There is, to-date, no therapy for this infection. Stx induces ATP release from host cells and ATP signaling mediates its cytotoxic effects. Apyrase cleaves and neutralizes ATP and its effect on Stx and EHEC infection was therefore investigated. Apyrase decreased bacterial RecA and dose-dependently decreased toxin release from E. coli O157:H7 in vitro, demonstrated by reduced phage DNA and protein levels. The effect was investigated in a mouse model of E. coli O157:H7 infection. BALB/c mice infected with Stx2-producing E. coli O157:H7 were treated with apyrase intraperitoneally, on days 0 and 2 post-infection, and monitored for 11 days. Apyrase-treated mice developed disease two days later than untreated mice. Untreated infected mice lost significantly more weight than those treated with apyrase. Apyrase-treated mice exhibited less colonic goblet cell depletion and apoptotic cells, as well as lower fecal ATP and Stx2, compared to untreated mice. Apyrase also decreased platelet aggregation induced by co-incubation of human platelet-rich-plasma with Stx2 and E. coli O157 lipopolysaccharide in the presence of collagen. Thus, apyrase had multiple protective effects, reducing RecA levels, stx2 and toxin release from EHEC, reducing fecal Stx2 and protecting mouse intestinal cells, as well as decreasing platelet activation, and could thereby delay the development of disease.


Subject(s)
Bacteriophages , Escherichia coli Infections , Escherichia coli O157 , Gastrointestinal Microbiome , Adenosine Triphosphate/metabolism , Animals , Apyrase/metabolism , Apyrase/pharmacology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Escherichia coli O157/genetics , Humans , Lipopolysaccharides/metabolism , Mice , Mice, Inbred BALB C , Shiga Toxin/metabolism , Shiga Toxin/pharmacology , Shiga Toxin 2/genetics , Shiga Toxin 2/metabolism , Shiga Toxin 2/pharmacology
2.
EMBO J ; 40(20): e107766, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34516001

ABSTRACT

The Golgi apparatus, the main glycosylation station of the cell, consists of a stack of discontinuous cisternae. Glycosylation enzymes are usually concentrated in one or two specific cisternae along the cis-trans axis of the organelle. How such compartmentalized localization of enzymes is achieved and how it contributes to glycosylation are not clear. Here, we show that the Golgi matrix protein GRASP55 directs the compartmentalized localization of key enzymes involved in glycosphingolipid (GSL) biosynthesis. GRASP55 binds to these enzymes and prevents their entry into COPI-based retrograde transport vesicles, thus concentrating them in the trans-Golgi. In genome-edited cells lacking GRASP55, or in cells expressing mutant enzymes without GRASP55 binding sites, these enzymes relocate to the cis-Golgi, which affects glycosphingolipid biosynthesis by changing flux across metabolic branch points. These findings reveal a mechanism by which a matrix protein regulates polarized localization of glycosylation enzymes in the Golgi and controls competition in glycan biosynthesis.


Subject(s)
Glycosphingolipids/metabolism , Golgi Apparatus/metabolism , Golgi Matrix Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Brefeldin A/pharmacology , Ceramides/metabolism , Cholera Toxin/pharmacology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Gene Expression , Glycosylation/drug effects , Golgi Apparatus/drug effects , Golgi Apparatus/genetics , Golgi Matrix Proteins/genetics , HeLa Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Shiga Toxin/pharmacology
3.
PLoS Pathog ; 17(2): e1009290, 2021 02.
Article in English | MEDLINE | ID: mdl-33529199

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) is a food-borne pathogen that causes diarrheal disease and the potentially lethal hemolytic uremic syndrome. We used an infant rabbit model of EHEC infection that recapitulates many aspects of human intestinal disease to comprehensively assess colonic transcriptional responses to this pathogen. Cellular compartment-specific RNA-sequencing of intestinal tissue from animals infected with EHEC strains containing or lacking Shiga toxins (Stx) revealed that EHEC infection elicits a robust response that is dramatically shaped by Stx, particularly in epithelial cells. Many of the differences in the transcriptional responses elicited by these strains were in genes involved in immune signaling pathways, such as IL23A, and coagulation, including F3, the gene encoding Tissue Factor. RNA FISH confirmed that these elevated transcripts were found almost exclusively in epithelial cells. Collectively, these findings suggest that Stx potently remodels the host innate immune response to EHEC.


Subject(s)
Colon/metabolism , Enterohemorrhagic Escherichia coli/physiology , Escherichia coli Infections/microbiology , Gene Expression Regulation , Intestinal Mucosa/metabolism , Shiga Toxin/pharmacology , Transcriptome/drug effects , Animals , Apoptosis , Colon/drug effects , Colon/pathology , Hemorrhage , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Rabbits
4.
Methods Mol Biol ; 2233: 53-70, 2021.
Article in English | MEDLINE | ID: mdl-33222127

ABSTRACT

Endocytosis and intracellular retrograde trafficking from endosomes to the Golgi apparatus are key cellular processes. Endocytosis is directly or indirectly involved in many if not all cellular functions ranging from nutrient uptake and receptor signaling to mitosis, cell division, and migration (Scita, Di Fiore. Nature 463(7280):464-473, 2010; McMahon, Boucrot. Nat Rev Mol Cell Biol 12(8):517-533, 2011). Retrograde trafficking is emerging as a key driver for cell polarity. Robust methods are needed to quantify these processes. At the example of the bacterial Shiga toxin and the endogenous α5ß1 integrin, we here describe generic methods to differentiate (1) internalized from cell surface-accessible cargo proteins and (2) endocytic cargo proteins that have reached the Golgi apparatus via the retrograde route from those that have not. The choice of antibodies or natural ligands allows to adjust these methods to virtually any chosen biological system.


Subject(s)
Endocytosis/genetics , Endosomes/genetics , Golgi Apparatus/genetics , Molecular Biology/methods , Biological Transport/genetics , Cell Movement/drug effects , Cell Polarity/genetics , HeLa Cells , Humans , Membrane Proteins/genetics , Metabolic Networks and Pathways/drug effects , Shiga Toxin/chemistry , Shiga Toxin/pharmacology , trans-Golgi Network
5.
Toxins (Basel) ; 13(1)2020 Dec 24.
Article in English | MEDLINE | ID: mdl-33374102

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) infections can cause EHEC-associated hemolytic uremic syndrome (eHUS) via its main virulent factor, Shiga toxins (Stxs). Complement has been reported to be involved in the progression of eHUS. The aim of this study was to investigate the interactions of the most effective subtype of the toxin, Stx2a, with pivotal complement proteins C3b and C5. The study further examined the effect of Stx2a stimulation on the transcription and synthesis of these complement proteins in human target cell lines. Binding of Stx2a to C3b and C5 was evaluated by ELISA. Kidney and gut cell lines (HK-2 and HCT-8) were stimulated with varied concentrations of Stx2a. Subsequent evaluation of complement gene transcription was studied by real-time PCR (qPCR), and ELISAs and Western blots were performed to examine protein synthesis of C3 and C5 in supernatants and lysates of stimulated HK-2 cells. Stx2a showed a specific binding to C3b and C5. Gene transcription of C3 and C5 was upregulated with increasing concentrations of Stx2a in both cell lines, but protein synthesis was not. This study demonstrates the binding of Stx2a to complement proteins C3b and C5, which could potentially be involved in regulating complement during eHUS infection, supporting further investigations into elucidating the role of complement in eHUS pathogenesis.


Subject(s)
Complement C3b/chemistry , Complement C5/chemistry , Gene Expression Regulation/drug effects , Shiga Toxin/chemistry , Shiga Toxin/pharmacology , Cell Line , Cell Survival , Humans , Protein Binding , Up-Regulation/drug effects
6.
J Biol Chem ; 295(13): 4341-4349, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32029474

ABSTRACT

Sphingolipid biosynthesis generates lipids for membranes and signaling that are crucial for many developmental and physiological processes. In some cases, large amounts of specific sphingolipids must be synthesized for specialized physiological functions, such as during axon myelination. How sphingolipid synthesis is regulated to fulfill these physiological requirements is not known. To identify genes that positively regulate membrane sphingolipid levels, here we employed a genome-wide CRISPR/Cas9 loss-of-function screen in HeLa cells using selection for resistance to Shiga toxin, which uses a plasma membrane-associated glycosphingolipid, globotriaosylceramide (Gb3), for its uptake. The screen identified several genes in the sphingolipid biosynthetic pathway that are required for Gb3 synthesis, and it also identified the aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor widely involved in development and physiology, as being required for Gb3 biosynthesis. AHR bound and activated the gene promoter of serine palmitoyltransferase small subunit A (SPTSSA), which encodes a subunit of the serine palmitoyltransferase that catalyzes the first and rate-limiting step in de novo sphingolipid biosynthesis. AHR knockout HeLa cells exhibited significantly reduced levels of cell-surface Gb3, and both AHR knockout HeLa cells and tissues from Ahr knockout mice displayed decreased sphingolipid content as well as significantly reduced expression of several key genes in the sphingolipid biosynthetic pathway. The sciatic nerve of Ahr knockout mice exhibited both reduced ceramide content and reduced myelin thickness. These results indicate that AHR up-regulates sphingolipid levels and is important for full axon myelination, which requires elevated levels of membrane sphingolipids.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Disease Resistance/genetics , Globosides/genetics , Receptors, Aryl Hydrocarbon/genetics , Serine C-Palmitoyltransferase/genetics , Sphingolipids/biosynthesis , Trihexosylceramides/genetics , Animals , CRISPR-Cas Systems/genetics , Gene Expression Regulation , Gene Knockout Techniques , Genome, Human/genetics , HeLa Cells , Humans , Lipid Metabolism/genetics , Lipids/biosynthesis , Lipids/genetics , Mice , Mice, Knockout , Shiga Toxin/pharmacology , Signal Transduction/genetics , Sphingolipids/genetics
7.
Soft Matter ; 16(6): 1614-1626, 2020 Feb 12.
Article in English | MEDLINE | ID: mdl-31957755

ABSTRACT

Membrane curvature effects are important in numerous cellular processes and many membrane interacting proteins induce spontaneous curvature upon membrane binding. Shiga and cholera toxins both belong to the AB5 family of toxins and consist of a toxic A subunit and a membrane-binding pentameric B subunit. Shiga and cholera toxins induce tubular membrane invaginations in cells and GUVs due to curvature effects and the toxins are known from MD simulations to induce curvature. Membrane invaginations have been linked to uptake of the toxins into cells. As a novel model system to experimentally characterize curvature-inducing proteins, we study the morphology induced in planar membrane patches. It was previously shown that annexins induce distinct morphologies in membrane patches including membrane rolling. In this study we show that the B subunits of Shiga and cholera toxins (STxB, CTxB) both induce roll-up of cell-sized membrane patches. Rolling starts from the free membrane edges of the patch and is completed within a few seconds. We characterize the branched roll morphology and find experimental estimates for the spontaneous curvature of the toxins based on the topography of rolls. The estimates are in agreement with previous MD simulations. We quantify the dynamics of rolling as induced by the toxins and demonstrate agreement with a theoretical model of the rolling dynamics. The model solves the equation of motion for a membrane roll and includes viscous drag and adhesion to the support. The results suggest that membrane rolling may be a general phenomenon displayed by many proteins that induce negative curvature in membranes with free edges.


Subject(s)
Cell Membrane/drug effects , Cholera Toxin/pharmacology , Molecular Dynamics Simulation , Shiga Toxin/pharmacology , Unilamellar Liposomes/chemistry , Cholera Toxin/chemistry , Shiga Toxin/chemistry
8.
Cell Rep ; 25(6): 1525-1536.e7, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30404007

ABSTRACT

The non-canonical caspase-4 and canonical NLRP3 inflammasomes are both activated by intracellular lipopolysaccharide (LPS), but the crosstalk between these two pathways remains unclear. Shiga toxin 2 (Stx2)/LPS complex, from pathogenic enterohemorrhagic Escherichia coli, activates caspase-4, gasdermin D (GSDMD), and the NLRP3 inflammasome in human THP-1 macrophages, but not mouse macrophages that lack the Stx receptor CD77. Stx2/LPS-mediated IL-1ß secretion and pyroptosis are dependent on mitochondrial reactive oxygen species (ROS) downstream of the non-canonical caspase-4 inflammasome and cleaved GSDMD, which is enriched at the mitochondria. Blockade of caspase-4 activation and ROS generation as well as GSDMD deficiency significantly reduces Stx2/LPS-induced IL-1ß production and pyroptosis. The NLRP3 inflammasome plays a significant role in amplifying Stx2/LPS-induced GSDMD cleavage and pyroptosis, with significant reduction of these responses in NLRP3-deficient THP-1 cells. Together, these data show that Stx2/LPS complex activates the non-canonical inflammasome and mitochondrial ROS upstream of the NLRP3 inflammasome to promote cytokine maturation and pyroptosis.


Subject(s)
Caspases, Initiator/metabolism , Inflammasomes/metabolism , Lipopolysaccharides/pharmacology , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neoplasm Proteins/metabolism , Reactive Oxygen Species/metabolism , Shiga Toxin/pharmacology , Animals , Cell Line , Enzyme Activation/drug effects , Humans , Intracellular Signaling Peptides and Proteins , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Mitochondria/drug effects , Phosphate-Binding Proteins , Pyroptosis/drug effects
9.
J Pharmacol Sci ; 138(1): 71-75, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30293960

ABSTRACT

Cancer remains a major health problem around the world. A Shiga toxin is a bacterial toxin often produced by Shigella dysenteriae and Escherichia coli. A subunit of the Shiga toxin (StxA) is a cytotoxic agent which could be used to induce death in cancer cells. StxA expressed from baculovirus was evaluated in a pTriEx™ expression vector. The baculovirus vector was used for the A subunit delivery of StxA. StxA cell cytotoxicity was induced by the virus and assessed in the MCF7 and HeLa cell lines. In addition, the breast cancer cytotoxicity of the expressed StxA was also assessed in a cancer induced in mice. The cytotoxicity of the recombinant StxA baculovirus with different multiplicities of infection (MOI) was measured. The results showed that significant cytotoxicity can be induced on the mammalian epithelial breast cancer cell lines, MCF7 and HeLa cells with MOI ≥ 2. The results also showed that a malignant tumor induced by MCF7 could be inhibited in a mouse cancer model. Therefore, it can be concluded that StxA, expressed by baculovirus, could be used for in vitro and in vivo gene delivery. In this study StxA, delivered by the baculovirus inhibited cell proliferation, and eliminated HeLa and MCF7 cells, in vitro. In conclusion, this method can be used as a safe alternative for anticancer drug delivery inside cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Drug Delivery Systems/methods , Escherichia coli , Gene Transfer Techniques , Shiga Toxin/pharmacology , Animals , Baculoviridae , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Disease Models, Animal , Genetic Vectors , Humans , Mice , Sf9 Cells
10.
ChemMedChem ; 11(22): 2506-2510, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27778487

ABSTRACT

Retro-1 is a small molecule that displays two important biological activities: First, it blocks the actions of certain toxins by altering their intracellular trafficking. Second, it enhances the activity of oligonucleotides by releasing them from entrapment in endosomes. This raises the question of whether the two actions involve the same cellular target. Herein we report the effects of several Retro-1 analogues on both toxins and oligonucleotides. We found analogues that affect toxins but not oligonucleotides and vice-versa, while Retro-1 is the only compound that affects both. This indicates that the molecular target(s) involved in the two processes are distinct.


Subject(s)
Benzodiazepinones/chemistry , Drug Delivery Systems , Oligonucleotides/chemistry , Shiga Toxin/pharmacology , Small Molecule Libraries/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Carriers/chemistry , HeLa Cells , Humans , Molecular Structure , Shiga Toxin/chemistry , Structure-Activity Relationship
11.
Sci Rep ; 6: 30336, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27458147

ABSTRACT

Shiga toxin (Stx), an AB5 toxin, binds specifically to the neutral glycosphingolipid Gb3 at the cell surface before being transported into cells. We here demonstrate that addition of conical lysophospholipids (LPLs) with large head groups inhibit Stx binding to cells whereas LPLs with small head groups do not. Lysophosphatidylinositol (LPI 18:0), the most efficient LPL with the largest head group, was selected for in-depth investigations to study how the binding of Stx is regulated. We show that the inhibition of Stx binding by LPI is reversible and possibly regulated by cholesterol since addition of methyl-ß-cyclodextrin (mßCD) reversed the ability of LPI to inhibit binding. LPI-induced inhibition of Stx binding is independent of signalling and membrane turnover as it occurs in fixed cells as well as after depletion of cellular ATP. Furthermore, data obtained with fluorescent membrane dyes suggest that LPI treatment has a direct effect on plasma membrane lipid packing with shift towards a liquid disordered phase in the outer leaflet, while lysophosphoethanolamine (LPE), which has a small head group, does not. In conclusion, our data show that cellular treatment with conical LPLs with large head groups changes intrinsic properties of the plasma membrane and modulates Stx binding to Gb3.


Subject(s)
Lysophospholipids/pharmacology , Neutral Glycosphingolipids/metabolism , Shiga Toxin/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , HeLa Cells , Humans , Lysophospholipids/chemistry , Protein Binding , beta-Cyclodextrins/pharmacology
12.
Cell Microbiol ; 18(10): 1339-48, 2016 10.
Article in English | MEDLINE | ID: mdl-26990252

ABSTRACT

Haemolytic anaemia is one of the characteristics of life-threatening extraintestinal complications in humans during infection with enterohaemorrhagic Escherichia coli (EHEC). Shiga toxins (Stxs) of EHEC preferentially damage microvascular endothelial cells of the kidney and the brain, whereby occluded small blood vessels may elicit anaemia through mechanical erythrocyte disruption. Here we show for the first time that Stx2a, the major virulence factor of EHEC, is also capable of direct targeting developing human erythrocytes. We employed an ex vivo erythropoiesis model using mobilized CD34(+) haematopoietic stem/progenitor cells from human blood and monitored expression of Stx receptors and Stx2a-mediated cellular injury of developing erythrocytes. CD34(+) haematopoietic stem/progenitor cells were negative for Stx2a receptors and resistant towards the toxin. Expression of Stx2a-binding glycosphingolipids and toxin sensitivity was apparent immediately after initiation of erythropoietic differentiation, peaked for basophilic and polychromatic erythroblast stages and declined during maturation into orthochromatic erythroblasts and reticulocytes, which became highly refractory to Stx2a. The observed Stx-mediated toxicity towards erythroblasts during the course of erythropoiesis might contribute, although speculative at this stage of research, to the anaemia caused by Stx-producing pathogens.


Subject(s)
Enterohemorrhagic Escherichia coli/physiology , Hematopoietic Stem Cells/physiology , Shiga Toxin/pharmacology , Cell Survival , Cells, Cultured , Erythrocytes/microbiology , Erythrocytes/physiology , Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/microbiology , Humans
13.
J Biomed Sci ; 22: 78, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26385393

ABSTRACT

BACKGROUND: In 2011, there was an outbreak of Shiga toxin-producing Escherichia coli (STEC) infections in Japan. Approximately 62 % of patients with hemolytic-uremic syndrome also showed symptoms of encephalopathy. To determine the mechanisms of onset for encephalopathy during STEC infections, we conducted an in vitro study with glial cell lines and primary glial cells. RESULTS: Shiga toxin 2 (Stx-2) in combination with lipopolysaccharide (LPS), or LPS alone activates nuclear factor-κB (NF-κB) signaling in glial cells. Similarly, Stx-2 in combination with LPS, or LPS alone increases expression levels of aquaporin 4 (AQP4) in glial cells. It is possible that overexpression of AQP4 results in a rapid and increased influx of osmotic water across the plasma membrane into cells, thereby inducing cell swelling and cerebral edema. CONCLUSIONS: We have showed that a combination of Stx-2 and LPS induced apoptosis of glial cells recently. Glial cells are indispensable for cerebral homeostasis; therefore, their dysfunction and death impairs cerebral homeostasis and results in encephalopathy. We postulate that the onset of encephalopathy in STEC infections occurs when Stx-2 attacks vascular endothelial cells of the blood-brain barrier, inducing their death. Stx-2 and LPS then attack the exposed glial cells that are no longer in contact with the endothelial cells. AQP4 is overexpressed in glial cells, resulting in their swelling and adversely affecting cerebral homeostasis. Once cerebral homeostasis is affected in such a way, encephalopathy is the likely result in STEC patients.


Subject(s)
Aquaporin 4/biosynthesis , Lipopolysaccharides/pharmacology , Neuroglia/metabolism , Shiga Toxin/pharmacology , Up-Regulation/drug effects , Animals , Cell Line , Rats
14.
Traffic ; 15(7): 772-87, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24703428

ABSTRACT

Several exogenous and endogenous cargo proteins are internalized independently of clathrin, including the bacterial Shiga toxin. The mechanisms underlying early steps of clathrin-independent uptake remain largely unknown. In this study, we have designed a protocol to obtain gradient fractions containing Shiga toxin internalization intermediates. Using stable isotope labeling with amino acids in cell culture (SILAC) and quantitative mass spectrometry, Rab12 was found in association with these very early uptake carriers. The localization of the GTPase on Shiga toxin-induced plasma membrane invaginations was shown by fluorescence microscopy in cells transfected with GFP-Rab12. Furthermore, using a quantitative biochemical assay, it was found that the amount of receptor-binding B-subunit of Shiga toxin reaching the trans-Golgi/TGN membranes was decreased in Rab12-depleted cells, and that cells were partially protected against intoxication by Shiga-like toxin 1 under these conditions. These findings demonstrate the functional importance of Rab12 for retrograde toxin trafficking. Among several other intracellular transport pathways, only the steady-state localizations of TGN46 and cation-independent mannose-6-phosphate receptor were affected. These data thus strongly suggest that Rab12 functions in the retrograde transport route.


Subject(s)
Shiga Toxin/pharmacology , rab GTP-Binding Proteins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Endocytosis , HeLa Cells , Humans , Protein Transport , Shiga Toxin/metabolism
15.
Blood ; 122(5): 803-6, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23733336

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) produce ribosome-inactivating Shiga toxins (Stx1, Stx2) responsible for development of hemolytic uremic syndrome (HUS) and acute kidney injury (AKI). Some patients show complement activation during EHEC infection, raising the possibility of therapeutic targeting of complement for relief. Our juvenile nonhuman primate (Papio baboons) models of endotoxin-free Stx challenge exhibit full spectrum HUS, including thrombocytopenia, hemolytic anemia, and AKI with glomerular thrombotic microangiopathy. There were no significant increases in soluble terminal complement complex (C5b-9) levels after challenge with lethal Stx1 (n = 6) or Stx2 (n = 5) in plasma samples from T0 to euthanasia at 49.5 to 128 hours post-challenge. d-dimer and cell injury markers (HMGB1, histones) confirmed coagulopathy and cell injury. Thus, complement activation is not required for the development of thrombotic microangiopathy and HUS induced by EHEC Shiga toxins in these preclinical models, and benefits or risks of complement inhibition should be studied further for this infection.


Subject(s)
Complement System Proteins/physiology , Hemolytic-Uremic Syndrome/immunology , Thrombotic Microangiopathies/immunology , Animals , Blood Coagulation/physiology , Complement Activation/physiology , Complement System Proteins/metabolism , Disease Models, Animal , Fibrin Fibrinogen Degradation Products/analysis , Hemolytic-Uremic Syndrome/complications , Hemolytic-Uremic Syndrome/etiology , Papio , Primates , Shiga Toxin/pharmacology , Shiga-Toxigenic Escherichia coli/pathogenicity , Shiga-Toxigenic Escherichia coli/physiology , Thrombotic Microangiopathies/complications , Thrombotic Microangiopathies/etiology , Thrombotic Microangiopathies/metabolism , Time Factors
16.
Tohoku J Exp Med ; 228(3): 247-52, 2012 11.
Article in English | MEDLINE | ID: mdl-23089636

ABSTRACT

Hemolytic uremic syndrome (HUS) in infants is mainly caused by the Shiga toxin (Stx), which is produced by pathogenic Escherichia coli O157:H7. Infants are prone to develop HUS in comparison to older children and adults, but its underlying mechanism remains unknown. Recent observations suggest that reactive oxygen species (ROS) and reactive nitrogen species (RNS) including nitric oxide (NO) may be involved in the pathogenesis of HUS. We therefore measured NO production by neutrophils prepared from infants (6-27 months old), children (5.3-11 years old) or adults (25-47 years old). The NO production was measured by a flow cytometric analysis with a fluorescent indicator (expressed as mean fluorescence intensity), and mRNA expression of inducible NO synthase (iNOS) was analyzed by reverse transcription-polymerase chain reaction (RT-PCR). The amount of NO produced was significantly lower in Stx-stimulated neutrophils prepared from infants (45.8 ± 23.3) than that in those from children (120.5 ± 81.5) or adults (127.7 ± 45.8) (n = 10 each group, P < 0.05). The expression level of iNOS mRNA was lower in Stx-stimulated neutrophils of the infants than the level in those of children or adults. In conclusion, Stx increased NO production in neutrophils probably via iNOS. Importantly, the degree of the Stx-mediated increase in NO production was lower in neutrophils of infants compared to those of children or adults, which may explain the higher incidence of HUS in infants. These results suggest that NO may contribute to the cellular defense mechanisms against Stx.


Subject(s)
Neutrophils/drug effects , Neutrophils/metabolism , Nitric Oxide/metabolism , Shiga Toxin/pharmacology , Adult , Aging/blood , Aging/genetics , Aging/immunology , Aging/metabolism , Cells, Cultured , Child , Child, Preschool , Gene Expression Regulation, Enzymologic/drug effects , Humans , Infant , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Middle Aged , Nitric Oxide/blood , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Shiga Toxin/immunology , Stimulation, Chemical
17.
Traffic ; 13(3): 443-54, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22132807

ABSTRACT

Shiga toxin inhibits protein synthesis after being transported from the cell surface to endosomes and retrogradely through the Golgi apparatus to the endoplasmic reticulum (ER) and into the cytosol. In this study, we have abolished proton gradients across internal membranes in different ways and investigated the effect on the various transport steps of Shiga toxin. Although inhibitors of the proton pump such as bafilomycin A1 and concanamycin A as well as some ionophores and chloroquine all protect against Shiga toxin, they mediate protection by inhibiting different transport steps. For instance, chloroquine protects the cells, although the toxin is transported to the ER. Importantly, our data indicate that proton pump activity is required for efficient endosome-to-Golgi transport of Shiga toxin, although acidification as such does not seem to be required.


Subject(s)
Endosomes/metabolism , Shiga Toxin/pharmacology , Animals , Biological Transport/drug effects , Cell Line , Chloroquine/pharmacology , Endoplasmic Reticulum/drug effects , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration , Macrolides/pharmacology , Nigericin/pharmacology
18.
PLoS One ; 5(7): e10944, 2010 Jul 27.
Article in English | MEDLINE | ID: mdl-20668539

ABSTRACT

Clathrin-dependent endocytosis is a main entry mechanism for the glycolipid-binding Shiga toxin (Stx), although clathrin-independent pathways are also involved. Binding of Stx to its receptor Gb3 not only is essential for Stx retrograde transport to the endoplasmic reticulum and toxicity but also activates signaling through the tyrosine kinase Syk. We previously described that Syk activity is important for Stx entry, but it remained unclear how this kinase modulates endocytosis of Stx. Here we characterized the effects of Stx and Syk on clathrin-coated pit formation. We found that acute treatment with Stx results in an increase in the number of clathrin-coated profiles as determined by electron microscopy and on the number of structures containing the endocytic AP-2 adaptor at the plasma membrane determined by live-cell spinning disk confocal imaging. These responses to Stx require functional Syk activity. We propose that a signaling pathway mediated by Syk and modulated by Stx leads to an increased number of endocytic clathrin-coated structures, thus providing a possible mechanism by which Stx enhances its own endocytosis.


Subject(s)
Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Coated Pits, Cell-Membrane/ultrastructure , Intracellular Signaling Peptides and Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Shiga Toxin/pharmacology , Adaptor Protein Complex 2/metabolism , Biological Transport/drug effects , Cell Line, Tumor , HeLa Cells , Humans , Microscopy, Electron , Syk Kinase
19.
Toxicon ; 56(4): 526-34, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20493201

ABSTRACT

We used two virtual screening programs, ICM and GOLD, to dock nearly 50,000 compounds into each of two conformations of the target protein ricin A chain (RTA). A limited control set suggests that candidates scored highly by two programs may have a higher probability of being ligands than those in a list from a single program. Based on the virtual screens, we purchased 306 compounds that were subjected to a kinetic assay. Six compounds were found to give modest, but significant, inhibition of RTA. They also tended to inhibit Shiga toxin A chain, with roughly the same IC(50). The compounds generally represent novel chemical platforms that do not resemble RTA substrates, as currently known inhibitors do. These six were also tested in a cell-based assay for their ability to protect cells from intact ricin. Two compounds were effective in this regard, showing modest to strong ricin inhibition, but also showing some cytotoxicity. RTA, with its large, polar active site is a difficult drug design target which is expected to bind small molecules only weakly. The ability of the method to find these novel platforms is encouraging and suggests virtual screening can contribute to the search for ricin and Shiga toxin inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Ricin/antagonists & inhibitors , Algorithms , Animals , Binding Sites , Chlorocebus aethiops , Enzyme Inhibitors/pharmacology , Inhibitory Concentration 50 , Kinetics , Libraries, Digital , Ligands , Molecular Conformation , Ricin/chemistry , Ricin/pharmacology , Shiga Toxin/antagonists & inhibitors , Shiga Toxin/chemistry , Shiga Toxin/pharmacology , Small Molecule Libraries , Software , Vero Cells
20.
Pediatr Nephrol ; 25(11): 2231-40, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20424866

ABSTRACT

Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli O157:H7 has become a global threat to public health, as a primary cause of a worldwide spread of hemorrhagic colitis complicated by diarrhea-associated hemolytic uremic syndrome (HUS), a disorder of thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure that mainly affects early childhood. Endothelial dysfunction has been recognized as the trigger event in the development of microangiopathic processes. Endothelial cells, mainly those located in the renal microvasculature, are primary targets of the toxic effects of Stx1 and 2. Stxs bound to their specific globotriaosylceramide (Gb3Cer) receptor on the cell surface trigger a cascade of signaling events, involving NF-κB activation, that induce expression of genes encoding for adhesion molecules and chemokines, and culminate in the adhesion of leukocytes to endothelial cells, thereby increasing the endothelial susceptibility to leukocyte-mediated injury. Activated endothelial cells in response to Stxs lose the normal thromboresistance phenotype and become thrombogenic, initiating microvascular thrombus formation. Evidence is emerging that complement activation in response to Stxs favors platelet thrombus formation on endothelial cells, which may play a role in amplifying the inflammation-thrombosis circuit in Stx-associated HUS.


Subject(s)
Endothelium, Vascular/physiopathology , Hemolytic-Uremic Syndrome/metabolism , Shiga Toxin/metabolism , Acute Kidney Injury/metabolism , Capillaries/metabolism , Chemokines/metabolism , Child, Preschool , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Escherichia coli O157/metabolism , Hemolytic-Uremic Syndrome/etiology , Hemolytic-Uremic Syndrome/physiopathology , Humans , Leukocytes/metabolism , Models, Biological , NF-kappa B/metabolism , Shiga Toxin/pharmacology , Trihexosylceramides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...