Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 222
Filter
1.
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
2.
Toxins (Basel) ; 15(3)2023 03 09.
Article in English | MEDLINE | ID: mdl-36977098

ABSTRACT

Shiga toxins (Stxs) produced by ingested E. coli can induce hemolytic uremic syndrome after crossing the intact intestinal barrier, entering the bloodstream, and targeting endothelial cells in the kidney. The method(s) by which the toxins reach the bloodstream are not fully defined. Here, we used two polarized cell models to evaluate Stx translocation: (i) a single-layer primary colonic epithelial cell model and (ii) a three-cell-layer model with colonic epithelial cells, myofibroblasts, and colonic endothelial cells. We traced the movement of Stx types 1a and 2a across the barrier models by measuring the toxicity of apical and basolateral media on Vero cells. We found that Stx1a and Stx2a crossed both models in either direction. However, approximately 10-fold more Stx translocated in the three-layer model as compared to the single-layer model. Overall, the percentage of toxin that translocated was about 0.01% in the epithelial-cell-only model but up to 0.09% in the three-cell-layer model. In both models, approximately 3- to 4-fold more Stx2a translocated than Stx1a. Infection of the three-cell-layer model with Stx-producing Escherichia coli (STEC) strains showed that serotype O157:H7 STEC reduced barrier function in the model and that the damage was not dependent on the presence of the eae gene. Infection of the three-layer model with O26:H11 STEC strain TW08571 (Stx1a+ and Stx2a+), however, allowed translocation of modest amounts of Stx without reducing barrier function. Deletion of stx2a from TW08571 or the use of anti-Stx1 antibody prevented translocation of toxin. Our results suggest that single-cell models may underestimate the amount of Stx translocation and that the more biomimetic three-layer model is suited for Stx translocation inhibitor studies.


Subject(s)
Escherichia coli Infections , Shiga-Toxigenic Escherichia coli , Animals , Chlorocebus aethiops , Shiga Toxin/metabolism , Vero Cells , Endothelial Cells/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Shiga Toxins/metabolism
3.
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
4.
Toxins (Basel) ; 12(9)2020 09 21.
Article in English | MEDLINE | ID: mdl-32967277

ABSTRACT

Many cattle are persistently colonized with Shiga toxin-producing Escherichia coli (STEC) and represent a major source of human infections with human-pathogenic STEC strains (syn. enterohemorrhagic E. coli (EHEC)). Intervention strategies most effectively protecting humans best aim at the limitation of bovine STEC shedding. Mechanisms enabling STEC to persist in cattle are only partialy understood. Cattle were long believed to resist the detrimental effects of Shiga toxins (Stxs), potent cytotoxins acting as principal virulence factors in the pathogenesis of human EHEC-associated diseases. However, work by different groups, summarized in this review, has provided substantial evidence that different types of target cells for Stxs exist in cattle. Peripheral and intestinal lymphocytes express the Stx receptor globotriaosylceramide (Gb3syn. CD77) in vitro and in vivo in an activation-dependent fashion with Stx-binding isoforms expressed predominantly at early stages of the activation process. Subpopulations of colonic epithelial cells and macrophage-like cells, residing in the bovine mucosa in proximity to STEC colonies, are also targeted by Stxs. STEC-inoculated calves are depressed in mounting appropriate cellular immune responses which can be overcome by vaccination of the animals against Stxs early in life before encountering STEC. Considering Stx target cells and the resulting effects of Stxs in cattle, which significantly differ from effects implicated in human disease, may open promising opportunities to improve existing yet insufficient measures to limit STEC carriage and shedding by the principal reservoir host.


Subject(s)
Cattle Diseases/microbiology , Escherichia coli Infections/veterinary , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Animals , Bacterial Shedding , Bacterial Zoonoses , Cattle , Cattle Diseases/immunology , Cattle Diseases/prevention & control , Cattle Diseases/transmission , Disease Vectors , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Escherichia coli Infections/transmission , Host-Pathogen Interactions , Shiga-Toxigenic Escherichia coli/immunology , Shiga-Toxigenic Escherichia coli/pathogenicity , Trihexosylceramides/metabolism , Virulence
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 Med Chem ; 63(15): 8114-8133, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32648758

ABSTRACT

High-throughput screening has shown that Retro-1 inhibits ricin and Shiga toxins by diminishing their intracellular trafficking via the retrograde route, from early endosomes to the Golgi apparatus. To improve the activity of Retro-1, a structure-activity relationship (SAR) study was undertaken and yielded an analogue with a roughly 70-fold better half-maximal effective concentration (EC50) against Shiga toxin cytotoxicity measured in a cell protein synthesis assay.


Subject(s)
Benzodiazepinones/chemistry , Benzodiazepinones/pharmacology , Shiga Toxins/antagonists & inhibitors , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , HeLa Cells , Humans , Protein Transport/drug effects , Protein Transport/physiology , Shiga Toxins/metabolism , Structure-Activity Relationship
7.
Sci Rep ; 10(1): 9752, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32546842

ABSTRACT

The two lectins LecA from Pseudomonas aeruginosa and the B-subunit of Shiga toxin from Shigella dysenteriae (StxB) share the glycosphingolipid globotriaosylceramide (Gb3) as receptor. Counterintuitively, we found that LecA and StxB segregated into different domains after recognizing Gb3 at the plasma membrane of cells. We hypothesized that the orientation of the carbohydrate head group of Gb3 embedded in the lipid bilayer differentially influences LecA and StxB binding. To test this hypothesis, we reconstituted lectin-Gb3 interaction using giant unilamellar vesicles and were indeed able to rebuild LecA and StxB segregation. Both, the Gb3 fatty acyl chain structure and the local membrane environment, modulated Gb3 recognition by LecA and StxB. Specifically, StxB preferred more ordered membranes compared to LecA. Based on our findings, we propose comparing staining patterns of LecA and StxB as an alternative method to assess membrane order in cells. To verify this approach, we re-established that the apical plasma membrane of epithelial cells is more ordered than the basolateral plasma membrane. Additionally, we found that StxB recognized Gb3 at the primary cilium and the periciliary membrane, whereas LecA only bound periciliary Gb3. This suggests that the ciliary membrane is of higher order than the surrounding periciliary membrane.


Subject(s)
Adhesins, Bacterial/metabolism , Protein Binding/physiology , Shiga Toxins/metabolism , Adhesins, Bacterial/physiology , Cell Membrane/metabolism , Epithelial Cells/metabolism , Glycosphingolipids/metabolism , Lectins/metabolism , Lectins/physiology , Ligands , Lipid Bilayers/chemistry , Protein Binding/genetics , Pseudomonas aeruginosa , Shiga Toxin/metabolism , Shigella dysenteriae , Trihexosylceramides/metabolism , Unilamellar Liposomes/metabolism
8.
Toxins (Basel) ; 12(6)2020 06 04.
Article in English | MEDLINE | ID: mdl-32512916

ABSTRACT

The global emergence of clinical diseases caused by enterohemorrhagic Escherichia coli (EHEC) is an issue of great concern. EHEC release Shiga toxins (Stxs) as their key virulence factors, and investigations on the cell-damaging mechanisms toward target cells are inevitable for the development of novel mitigation strategies. Stx-mediated hemolytic uremic syndrome (HUS), characterized by the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute renal injury, is the most severe outcome of an EHEC infection. Hemolytic anemia during HUS is defined as the loss of erythrocytes by mechanical disruption when passing through narrowed microvessels. The formation of thrombi in the microvasculature is considered an indirect effect of Stx-mediated injury mainly of the renal microvascular endothelial cells, resulting in obstructions of vessels. In this review, we summarize and discuss recent data providing evidence that HUS-associated hemolytic anemia may arise not only from intravascular rupture of erythrocytes, but also from the extravascular impairment of erythropoiesis, the development of red blood cells in the bone marrow, via direct Stx-mediated damage of maturing erythrocytes, leading to "non-hemolytic" anemia.


Subject(s)
Erythrocytes/microbiology , Erythropoiesis , Escherichia coli Infections/microbiology , Hemolytic-Uremic Syndrome/microbiology , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Animals , Down-Regulation , Erythrocytes/metabolism , Erythrocytes/pathology , Escherichia coli Infections/blood , Escherichia coli Infections/pathology , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/pathology , Host-Pathogen Interactions , Humans , Shiga Toxins/blood , Shiga-Toxigenic Escherichia coli/pathogenicity , Stress, Mechanical
9.
Toxins (Basel) ; 12(5)2020 05 22.
Article in English | MEDLINE | ID: mdl-32456007

ABSTRACT

Shiga toxin (STx) produced by Shigella and closely related Shiga toxin 1 and 2 (STx1 and STx2) synthesized by Shiga toxin-producing Escherichia coli (STEC) are bacterial AB5 toxins. All three toxins target kidney cells and may cause life-threatening renal disease. While Shigella infections can be treated with antibiotics, resistance is increasing. Moreover, antibiotic therapy is contraindicated for STEC, and there are no definitive treatments for STEC-induced disease. To exert cellular toxicity, STx, STx1, and STx2 must undergo retrograde trafficking to reach their cytosolic target, ribosomes. Direct transport from early endosomes to the Golgi apparatus is an essential step that allows the toxins to bypass degradative late endosomes and lysosomes. The essentiality of this transport step also makes it an ideal target for the development of small-molecule inhibitors of toxin trafficking as potential therapeutics. Here, we review the recent advances in understanding the molecular mechanisms of the early endosome-to-Golgi transport of STx, STx1, and STx2, as well as the development of small-molecule inhibitors of toxin trafficking that act at the endosome/Golgi interface.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Dysentery, Bacillary/drug therapy , Endosomes/metabolism , Escherichia coli Infections/drug therapy , Golgi Apparatus/metabolism , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/drug effects , Shigella/drug effects , Animals , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/microbiology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Humans , Molecular Targeted Therapy , Protein Transport , Shiga-Toxigenic Escherichia coli/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity , Shigella/metabolism , Shigella/pathogenicity
10.
Toxins (Basel) ; 12(5)2020 05 23.
Article in English | MEDLINE | ID: mdl-32456125

ABSTRACT

Shiga toxins (Stxs), syn. Vero(cyto)toxins, are potent bacterial exotoxins and the principal virulence factor of enterohemorrhagic Escherichia coli (EHEC), a subset of Shiga toxin-producing E. coli (STEC). EHEC strains, e.g., strains of serovars O157:H7 and O104:H4, may cause individual cases as well as large outbreaks of life-threatening diseases in humans. Stxs primarily exert a ribotoxic activity in the eukaryotic target cells of the mammalian host resulting in rapid protein synthesis inhibition and cell death. Damage of endothelial cells in the kidneys and the central nervous system by Stxs is central in the pathogenesis of hemolytic uremic syndrome (HUS) in humans and edema disease in pigs. Probably even more important, the toxins also are capable of modulating a plethora of essential cellular functions, which eventually disturb intercellular communication. The review aims at providing a comprehensive overview of the current knowledge of the time course and the consecutive steps of Stx/cell interactions at the molecular level. Intervention measures deduced from an in-depth understanding of this molecular interplay may foster our basic understanding of cellular biology and microbial pathogenesis and pave the way to the creation of host-directed active compounds to mitigate the pathological conditions of STEC infections in the mammalian body.


Subject(s)
Escherichia coli Infections/microbiology , Hemolytic-Uremic Syndrome/microbiology , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Virulence Factors/metabolism , Animals , Apoptosis , Escherichia coli Infections/metabolism , Escherichia coli Infections/pathology , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/pathology , Host-Pathogen Interactions , Humans , Shiga-Toxigenic Escherichia coli/pathogenicity , Signal Transduction
11.
Cell Mol Gastroenterol Hepatol ; 10(1): 171-190, 2020.
Article in English | MEDLINE | ID: mdl-32145469

ABSTRACT

BACKGROUND & AIMS: Shiga toxin (Stx)-producing Escherichia coli (eg, O157:H7) infection produces bloody diarrhea, while Stx inhibits protein synthesis and causes the life-threatening systemic complication of hemolytic uremic syndrome. The murine intestinal tract is resistant to O157:H7 and Stx, and human cells in culture fail to model the complex tissue responses to intestinal injury. We used genetically identical, human stem cell-derived intestinal tissues of varying complexity to study Stx toxicity in vitro and in vivo. METHODS: In vitro susceptibility to apical or basolateral exposure to Stx was assessed using human intestinal organoids (HIOs) derived from embryonic stem cells, or enteroids derived from multipotent intestinal stem cells. HIOs contain a lumen, with a single layer of differentiated epithelium surrounded by mesenchymal cells. Enteroids only contain epithelium. In vivo susceptibility was assessed using HIOs, with or without an enteric nervous system, transplanted into mice. RESULTS: Stx induced necrosis and apoptotic death in both epithelial and mesenchymal cells. Responses that require protein synthesis (cellular proliferation and wound repair) also were observed. Epithelial barrier function was maintained even after epithelial cell death was seen, and apical to basolateral translocation of Stx was seen. Tissue cross-talk, in which mesenchymal cell damage caused epithelial cell damage, was observed. Stx induced mesenchymal expression of the epithelial marker E-cadherin, the initial step in mesenchymal-epithelial transition. In vivo responses of HIO transplants injected with Stx mirrored those seen in vitro. CONCLUSIONS: Intestinal tissue responses to protein synthesis inhibition by Stx are complex. Organoid models allow for an unprecedented examination of human tissue responses to a deadly toxin.


Subject(s)
Epithelial Cells/pathology , Escherichia coli Infections/pathology , Hemolytic-Uremic Syndrome/pathology , Shiga Toxins/toxicity , Animals , Apoptosis , Cell Line , Disease Models, Animal , Escherichia coli Infections/microbiology , Hemolytic-Uremic Syndrome/microbiology , Human Embryonic Stem Cells , Humans , Intestinal Mucosa , Mice , Necrosis , Organoids , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity
12.
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
13.
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
14.
Microbiologyopen ; 8(2): e00636, 2019 02.
Article in English | MEDLINE | ID: mdl-29675935

ABSTRACT

Temperate phage encoded Shiga toxin (Stx) kills the bacterivorous predator, Tetrahymena thermophila, providing Stx+ Escherichia coli with a survival advantage over Stx- cells. Although bacterial death accompanies Stx release, since bacteria grow clonally the fitness benefits of predator killing accrue to the kin of the sacrificed organism, meaning Stx-mediated protist killing is a form of self-destructive cooperation. We show here that the fitness benefits of Stx production are not restricted to the kin of the phage-encoding bacteria. Instead, nearby "free loading" bacteria, irrespective of their genotype, also reap the benefit of Stx-mediated predator killing. This finding indicates that the phage-borne Stx exotoxin behaves as a public good. Stx is encoded by a mobile phage. We find that Stx-encoding phage can use susceptible bacteria in the population as surrogates to enhance toxin and phage production. Moreover, our findings also demonstrate that engulfment and concentration of Stx-encoding and susceptible Stx- bacteria in the Tetrahymena phagosome enhances the transfer of Stx-encoding temperate phage from the host to the susceptible bacteria. This transfer increases the population of cooperating bacteria within the community. Since these bacteria now encode Stx, the predation-stimulated increase in phage transfer increases the population of toxin encoding bacteria in the environment.


Subject(s)
Antibiosis , Coliphages/genetics , Escherichia coli/growth & development , Escherichia coli/virology , Shiga Toxins/toxicity , Tetrahymena thermophila/drug effects , Tetrahymena thermophila/growth & development , Microbial Interactions , Shiga Toxins/genetics , Shiga Toxins/metabolism
15.
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
16.
Int J Food Microbiol ; 257: 201-205, 2017 Sep 18.
Article in English | MEDLINE | ID: mdl-28672173

ABSTRACT

Verocytotoxin (VT)-producing Escherichia coli (VTEC) are a significant foodborne public health hazard, where most human infections are associated with six serogroups (O157, O26, O103, O145, O111 and O104). VTEC was the fourth most commonly reported zoonosis in the EU in 2015, with 5901 confirmed human cases. Ruminant animals, including cattle, are a major reservoir of VTEC. The consumption of VTEC-contaminated animal-derived foodstuffs, especially undercooked ground beef, is an important transmission route. To the best of our knowledge, there are few data available on the contamination of VTEC in meat products in Italy. During 2015 and 2016, 250 raw meat samples were collected from retail markets in southern Italy (Apulia) and analysed for the occurrence of vtx genes (vtx1/vtx2) at the Istituto Zooprofilattico Sperimentale della Puglia e della Basilicata (IZS PB, Italy). In addition, the isolates were characterized by determining the presence of VTEC main virulence factors, the antimicrobial resistance profiles and the genetic relatedness by pulsed-field gel electrophoresis (PFGE). The results have shown that 8.4% (21/250) of the samples were positive for vtx genes in the preliminary screening step but VTEC strains were isolated from only 2% (5/250) of overall meat analysed samples, including raw ground beef, beef hamburger and beef carpaccio. 5 isolates displayed a multi-drug resistance phenotype. All VTEC strains were analysed by XbaI-PFGE and dendrogram revealed 5 distinct restriction profiles, indicating their relatively high genetic diversity. Although this study demonstrates a low prevalence of VTEC in raw beef marketed in southern Italy, the presence of potentially pathogenic E. coli strains points to the need for proper hygiene during meat production to reduce the risk of foodborne illness and transmission of multi-drug resistant organisms via foods to humans.


Subject(s)
Meat Products/microbiology , Red Meat/microbiology , Shiga Toxins/genetics , Shiga-Toxigenic Escherichia coli/genetics , Shiga-Toxigenic Escherichia coli/isolation & purification , Animals , Cattle , Drug Resistance, Multiple, Bacterial/genetics , Electrophoresis, Gel, Pulsed-Field , Food Safety , Foodborne Diseases/microbiology , Humans , Italy , Prevalence , Shiga Toxins/metabolism , Shiga-Toxigenic Escherichia coli/drug effects
17.
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
18.
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
19.
Chem Biol Interact ; 267: 96-103, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-27712998

ABSTRACT

Medical countermeasures to treat biothreat agent infections require broad-spectrum therapeutics that do not induce agent resistance. A cell-based high-throughput screen (HTS) against ricin toxin combined with hit optimization allowed selection of a family of compounds that meet these requirements. The hit compound Retro-2 and its derivatives have been demonstrated to be safe in vivo in mice even at high doses. Moreover, Retro-2 is an inhibitor of retrograde transport that affects syntaxin-5-dependent toxins and pathogens. As a consequence, it has a broad-spectrum activity that has been demonstrated both in vitro and in vivo against ricin, Shiga toxin-producing O104:H4 entero-hemorrhagic E. coli and Leishmania sp. and in vitro against Ebola, Marburg and poxviruses and Chlamydiales. An effect is anticipated on other toxins or pathogens that use retrograde trafficking and syntaxin-5. Since Retro-2 targets cell components of the host and not directly the pathogen, no selection of resistant pathogens is expected. These lead compounds need now to be developed as drugs for human use.


Subject(s)
Benzamides/pharmacology , Chlamydiales/metabolism , Ebolavirus/metabolism , Leishmania/metabolism , Ricin/metabolism , Shiga Toxins/metabolism , Thiophenes/pharmacology , Animals , Benzamides/chemistry , Body Weight/drug effects , Chlamydiales/drug effects , Ebolavirus/drug effects , Escherichia coli/metabolism , HEK293 Cells , HeLa Cells , Humans , Injections, Intraperitoneal , Leishmania/drug effects , Mice , Mice, Inbred BALB C , Mitomycin/pharmacology , Models, Animal , RAW 264.7 Cells , Ricin/antagonists & inhibitors , Shiga Toxins/antagonists & inhibitors , Thiophenes/chemistry
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...