Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Appl Environ Microbiol ; 89(4): e0130622, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37014232

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) O157:H7 is a foodborne pathogen producing Shiga toxins (Stx1 and Stx2), which can cause hemorrhagic diarrhea and life-threatening infections. O157:H7 strain EDL933 carries prophages CP-933V and BP-933W, which encode Shiga toxin genes (stx1 and stx2, respectively). The aim of this work was to investigate the mechanisms of adaptive resistance of EHEC strain EDL933 to a typically lethal dose of gamma irradiation (1.5 kGy). Adaptive selection through six passages of exposure to 1.5 kGy resulted in the loss of CP-933V and BP-933W prophages from the genome and mutations within three genes: wrbA, rpoA, and Wt_02639 (molY). Three selected EHEC clones that became irradiation adapted to the 1.5-kGy dose (C1, C2, and C3) demonstrated increased resistance to oxidative stress, sensitivity to acid pH, and decreased cytotoxicity to Vero cells. To confirm that loss of prophages plays a role in increased radioresistance, clones C1 and C2 were exposed to bacteriophage-containing lysates. Although phage BP-933W could lysogenize C1, C2, and E. coli K-12 strain MG1655, it was not found to have integrated into the bacterial chromosome in C1-Φ and C2-Φ lysogens. Interestingly, for the E. coli K-12 lysogen (K-12-Φ), BP-933W DNA had integrated at the wrbA gene (K-12-Φ). Both C1-Φ and C2-Φ lysogens regained sensitivity to oxidative stress, were more effectively killed by a 1.5-kGy gamma irradiation dose, and had regained cytotoxicity and acid resistance phenotypes. Further, the K-12-Φ lysogen became cytotoxic, more sensitive to gamma irradiation and oxidative stress, and slightly more acid resistant. IMPORTANCE Gamma irradiation of food products can provide an effective means of eliminating bacterial pathogens such as enterohemorrhagic Escherichia coli (EHEC) O157:H7, a significant foodborne pathogen that can cause severe disease due to the production of Stx. To decipher the mechanisms of adaptive resistance of the O157:H7 strain EDL933, we evolved clones of this bacterium resistant to a lethal dose of gamma irradiation by repeatedly exposing bacterial cells to irradiation following a growth restoration over six successive passages. Our findings provide evidence that adaptive selection involved modifications in the bacterial genome, including deletion of the CP-933V and BP-933W prophages. These mutations in EHEC O157:H7 resulted in loss of stx1 and stx2, loss of cytotoxicity to epithelial cells, and decreased resistance to acidity, critical virulence determinants of EHEC, concomitant with increased resistance to lethal irradiation and oxidative stress. These findings demonstrate that the potential adaptation of EHEC to high doses of radiation would involve elimination of the Stx-encoding phages and likely lead to a substantial attenuation of virulence.


Subject(s)
Bacteriophages , Enterohemorrhagic Escherichia coli , Escherichia coli O157 , Escherichia coli Proteins , Animals , Chlorocebus aethiops , Shiga Toxin/genetics , Prophages/genetics , Vero Cells , Shiga Toxins/pharmacology , Bacteriophages/genetics , Genomics , Repressor Proteins/pharmacology
2.
Asian Pac J Cancer Prev ; 23(10): 3247-3252, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36308345

ABSTRACT

BACKGROUND: The low general toxicity against tumors expressing globotriaosylceramide (Gb3) and Shiga-like toxins produced by E. coli have been proposed as an anti-cancer therapy because of their specific target. This study aimed to determine the potency of the local strains of E. coli O157:H7 isolated from humans and cattle as a new breast cancer therapy by analyzing the cell cycle's inhibition and apoptosis induction. MATERIAL AND METHODS: Approximately 10 cultured T47D cells were subjected to Shiga-like toxin produced by four local isolates of E. coli O157:H7, including KL-48 (2) from humans, and SM-25 (1), SM-7 (1), DS-21 (4) from cattle. Using ATCC 43894 as a control, the treatment was observed for 24 h by two replications. In addition, a FITC-Annexin V and PI assay were used to observe apoptosis and necrosis effect, as well as to analyze the cell cycle using propidium iodide (PI) staining. RESULTS: The results showed the toxicity effect of Shiga in the human T47 D cells line. The viability of the cells is subjected to Shiga-like toxins produced by KL-48 (2), SM7 (1), ATCC 43894, SM-25 (1), and DS-21 (4) isolates decreased with 15.20, 16.36, 22.17,  22.64, and 33.86%, in contrary to control of 94.36%. These were supported by the cells entering the late apoptosis of the cell cycle through each isolate with 67.66, 62.60, 63.68, 63.90, and 54.74%, and a control of 0.01%. Also, the necrosis cell for each treatment of 12.73, 19.3, 10.84, 10.53, and 4.86% was higher than the control of 5.51%. These were confirmed by the higher percentage of the cells treated with toxins of KL-48 (2), SM7(1), ATCC 43894, SM-25 (1), and DS-21 (4), which entered G0-G1 of the cell cycle phase with 66.41, 63.37, 61.52, 55.36, and 47.28%, respectively, than control of 40.69%. Additionally, the toxicity effect was supported by an increase in the cells entering the S and the G2-M phase of the cycle for each treatment. CONCLUSION: It is concluded that the Shiga-like toxin produced by E. coli O157:H7 local isolates can be developed as a drug against breast cancer based on its effect to arrest induction of the cell cycle and inducing apoptosis.


Subject(s)
Breast Neoplasms , Escherichia coli Infections , Escherichia coli O157 , Cattle , Humans , Animals , Female , Flow Cytometry , Breast Neoplasms/drug therapy , Shiga Toxins/genetics , Shiga Toxins/pharmacology , Shiga Toxins/therapeutic use , Cell Division , Cell Cycle , Apoptosis , Necrosis , Escherichia coli Infections/drug therapy
3.
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
4.
Eur J Pharmacol ; 899: 174057, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33753109

ABSTRACT

Immunotoxins are protein-based drugs consist of a target-specific binding domain and a cytotoxic domain to eliminate target cells. Such compounds are potentially therapeutic to combat diseases such as cancer. Generally, the B-subunit of Shiga toxin (STXB) receptor, globotriaosylceramide (Gb3), is expressed in high amounts on a number of human tumors cancer cells. In this study, we evaluated a new antitumor candidate called DT389-STXB chimeric protein, which genetically fused the DT to B-subunit of Shiga-like toxin (STXB). First a chimeric protein, encoding DT389-STXB was synthesized. The optimized chimeric protein expressed in E.coli BL21 (DE3) and confirmed by anti-His Western blot analysis. T47D, SKBR3, 4T1 and MCF7 cell lines were treated separately with purified DT389-STXB recombinant protein and functional activity of DT389-STXB was analyzed by the cell enzyme-linked immunosorbentassay (ELISA), MTT, ICC, Western blot and apoptosis tests. The results indicated that the recombinant DT389-STXB fusion protein with a molecular weight of 53 kDa was successfully expressed in E.coli BL21 (DE3) and the anti-His western-blot was used to confirm the presence of the protein. The DT389-STXB fusion protein attached to T47D, SKBR3 and 4T1 cell lines with the proper affinity and induced dose-dependent cytotoxicity against GB3-expressing cancer cells in vitro. Our results showed that DT389-STXB fusion protein may be a promising candidate for antitumor therapy agent against breast cancer; however, further studies are required to explore its efficacy in vivo for therapeutic applications.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Diphtheria Toxin/pharmacology , Shiga Toxins/pharmacology , Animals , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Female , Humans , MCF-7 Cells , Mice , Recombinant Fusion Proteins/pharmacology , Trihexosylceramides/metabolism
5.
FEBS Lett ; 594(15): 2406-2420, 2020 08.
Article in English | MEDLINE | ID: mdl-32473599

ABSTRACT

The interaction between the Shiga toxin B-subunit (STxB) and its globotriaosylceramide receptor (Gb3) has a high potential for being exploited for targeted cancer therapy. The primary goal of this study was to evaluate the capacity of STxB to carry small molecules and proteins as cargo into cells. For this purpose, an assay was designed to provide real-time information about the StxB-Gb3 interaction as well as the dynamics and mechanism of the internalization process. The assay revealed the ability to distinguish the process of binding to the cell surface from internalization and presented the importance of receptor and STxB clustering for internalization. The overall setup demonstrated that the binding mechanism is complex, and the concept of affinity is difficult to apply. Hence, time-resolved methods, providing detailed information about the interaction of STxB with cells, are critical for the optimization of intracellular delivery.


Subject(s)
Biological Assay , Drug Carriers , Neoplasms/metabolism , Shiga Toxins , Trihexosylceramides/metabolism , Biological Transport, Active , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , HT29 Cells , Humans , K562 Cells , Neoplasms/drug therapy , Neoplasms/pathology , Shiga Toxins/pharmacokinetics , Shiga Toxins/pharmacology
6.
Oncogene ; 37(18): 2410-2421, 2018 05.
Article in English | MEDLINE | ID: mdl-29440708

ABSTRACT

The globotriaosylceramide Gb3 is a glycosphingolipid expressed on a subpopulation of germinal center B lymphocytes which has been recognized as the B cell differentiation antigen CD77. Among tumoral cell types, Gb3/CD77 is strongly expressed in Burkitt's lymphoma (BL) cells as well as other solid tumors including breast, testicular and ovarian carcinomas. One known ligand of Gb3/CD77 is Verotoxin-1 (VT-1), a Shiga toxin produced in specific E. coli strains. Previously, we have reported that in BL cells, VT-1 induces apoptosis via a caspase-dependent and mitochondria-dependent pathway. Yet, the respective roles of various apoptogenic factors remained to be deciphered. Here, this apoptotic pathway was found to require cleavage of the BID protein by caspase-8 as well as activation of two other apoptogenic proteins, BAK and BAX. Surprisingly however, t-BID, the truncated form of BID resulting from caspase-8 cleavage, played no role in the conformational changes of BAK and BAX. Rather, their activation occurred under the control of full length BID (FL-BID). Indeed, introducing a non-cleavable form of BID (BID-D59A) into BID-deficient BL cells restored BAK and BAX activation following VT-1 treatment. Still, t-BID was involved along with FL-BID in the BAK-dependent and BAX-dependent cytosolic release of CYT C and SMAC/DIABLO from the mitochondrial intermembrane space: FL-BID was found to control the homo-oligomerization of both BAK and BAX, likely contributing to the initial release of CYT C and SMAC/DIABLO, while t-BID was needed for their hetero-oligomerization and ensuing release amplification. Together, our results reveal a functional cooperation between BAK and BAX during VT-1-induced apoptosis and, unexpectedly, that activation of caspase-8 and production of t-BID were not mandatory for initiation of the cell death process.


Subject(s)
Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/physiology , Burkitt Lymphoma/pathology , Shiga Toxins/pharmacology , Apoptosis/genetics , BH3 Interacting Domain Death Agonist Protein/chemistry , BH3 Interacting Domain Death Agonist Protein/genetics , Burkitt Lymphoma/genetics , Caspase 8/metabolism , HEK293 Cells , Humans , Protein Domains/genetics , Protein Domains/physiology , Protein Isoforms/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Cells, Cultured , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/physiology
7.
Methods Mol Biol ; 1571: 233-249, 2017.
Article in English | MEDLINE | ID: mdl-28281260

ABSTRACT

To improve food safety there is a need to develop simple, low-cost sensitive devices for detection of food-borne pathogens and their toxins. We describe a simple, low-cost webcam-based detector which can be used for various optical detection modalities, including fluorescence, chemiluminescence, densitometry, and colorimetric assays. The portable battery-operated CCD-based detection system consists of four modules: (1) a webcam to measure and record light emission, (2) a sample plate to perform assays, (3) a light emitting diode (LED) for illumination, and (4) a portable computer to acquire and analyze images. To demonstrate the technology, we used a cell based assay for fluorescence detection of the activity of the food borne Shiga toxin type 2 (Stx2), differentiating between biologically active toxin and inactive toxin which is not a risk. The assay is based on Shiga toxin inhibition of cell protein synthesis measured through inhibition of the green fluorescent protein (GFP). In this assay, GFP emits light at 509 nm when excited with a blue LED equipped with a filter at 486 nm. The emitted light is then detected with a green filter at 535 nm. Toxin activity is measured through a reduction in the 509 nm emission. In this system the level of detection (LOD) for Stx2 was 0.1 pg/ml, similar to the LOD of commercial fluorometers. These results demonstrate the utility and potential of low cost detectors for toxin activity. This approach could be readily adapted to the detection of other food-borne toxins.


Subject(s)
Biosensing Techniques/instrumentation , Biosensing Techniques/methods , Optical Devices , Shiga Toxins , Adenoviridae/genetics , Animals , Biological Assay , Cell Culture Techniques , Cell Line , Gene Expression , Genes, Reporter , Genetic Vectors/genetics , Humans , Image Processing, Computer-Assisted/methods , Molecular Imaging/instrumentation , Molecular Imaging/methods , Shiga Toxins/pharmacology , Statistics as Topic/methods , Transduction, Genetic
8.
PLoS One ; 11(12): e0166948, 2016.
Article in English | MEDLINE | ID: mdl-27935997

ABSTRACT

Many germ line diseases stem from a relatively minor disturbance in mutant protein endoplasmic reticulum (ER) 3D assembly. Chaperones are recruited which, on failure to correct folding, sort the mutant for retrotranslocation and cytosolic proteasomal degradation (ER-associated degradation-ERAD), to initiate/exacerbate deficiency-disease symptoms. Several bacterial (and plant) subunit toxins, retrograde transport to the ER after initial cell surface receptor binding/internalization. The A subunit has evolved to mimic a misfolded protein and hijack the ERAD membrane translocon (dislocon), to effect cytosolic access and cytopathology. We show such toxins compete for ERAD to rescue endogenous misfolded proteins. Cholera toxin or verotoxin (Shiga toxin) containing genetically inactivated (± an N-terminal polyleucine tail) A subunit can, within 2-4 hrs, temporarily increase F508delCFTR protein, the major cystic fibrosis (CF) mutant (5-10x), F508delCFTR Golgi maturation (<10x), cell surface expression (20x) and chloride transport (2x) in F508del CFTR transfected cells and patient-derived F508delCFTR bronchiolar epithelia, without apparent cytopathology. These toxoids also increase glucocerobrosidase (GCC) in N370SGCC Gaucher Disease fibroblasts (3x), another ERAD-exacerbated misfiling disease. We identify a new, potentially benign approach to the treatment of certain genetic protein misfolding diseases.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/drug effects , Endoplasmic Reticulum/drug effects , Protein Folding/drug effects , Toxins, Biological/pharmacology , Blotting, Western , Cholera Toxin/pharmacology , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/prevention & control , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Microscopy, Fluorescence , Models, Biological , Mutation , Protein Transport/drug effects , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/prevention & control , Shiga Toxins/pharmacology , Toxins, Biological/classification
9.
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
10.
Mol Cancer Ther ; 15(5): 1008-17, 2016 05.
Article in English | MEDLINE | ID: mdl-26826119

ABSTRACT

The B-subunit of the bacterial Shiga toxin (STxB), which is nontoxic and has low immunogenicity, can be used for tumor targeting of breast, colon, and pancreatic cancer. Here, we tested whether human gastric cancers, which are among the most aggressive tumor entities, express the cellular receptor of Shiga toxin, the glycosphingolipid globotriaosylceramide (Gb3/CD77). The majority of cases showed an extensive staining for Gb3 (36/50 cases, 72%), as evidenced on tissue sections of surgically resected specimen. Gb3 expression was detected independent of type (diffuse/intestinal), and was negatively correlated to increasing tumor-node-metastasis stages (P = 0.0385), as well as with markers for senescence. Gb3 expression in nondiseased gastric mucosa was restricted to chief and parietal cells at the bottom of the gastric glands, and was not elevated in endoscopic samples of gastritis (n = 10). Gb3 expression in established cell lines of gastric carcinoma was heterogeneous, with 6 of 10 lines being positive, evidenced by flow cytometry. STxB was taken up rapidly by live Gb3-positive gastric cancer cells, following the intracellular retrograde transport route, avoiding lysosomes and rapidly reaching the Golgi apparatus and the endoplasmic reticulum. Treatment of the Gb3-expressing gastric carcinoma cell line St3051 with STxB coupled to SN38, the active metabolite of the topoisomerase type I inhibitor irinotecan, resulted in >100-fold increased cytotoxicity, as compared with irinotecan alone. No cytotoxicity was observed on gastric cancer cell lines lacking Gb3 expression, demonstrating receptor specificity of the STxB-SN38 compound. Thus, STxB is a highly specific transport vehicle for cytotoxic agents in gastric carcinoma. Mol Cancer Ther; 15(5); 1008-17. ©2016 AACR.


Subject(s)
Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Immunotoxins/pharmacology , Shiga Toxins/pharmacology , Stomach Neoplasms/metabolism , Trihexosylceramides/metabolism , Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Animals , Biomarkers , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cellular Senescence/genetics , Dose-Response Relationship, Drug , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gene Expression , Humans , Molecular Targeted Therapy , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Trihexosylceramides/genetics
11.
J Microbiol Biotechnol ; 26(2): 432-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26643967

ABSTRACT

Shiga toxins (Stxs) produced by Shiga toxin-producing Escherichia coli (STEC) strains are major virulence factors that cause fatal systemic complications, such as hemolytic uremic syndrome and disruption of the central nervous system. Although numerous studies report proinflammatory responses to Stx type 1 (Stx1) or Stx type 2 (Stx2) both in vivo and in vitro, none have examined dynamic immune regulation involving cytokines and/or unknown inflammatory mediators during intoxication. Here, we showed that enzymatically active Stxs trigger the dissociation of lysyl-tRNA synthetase (KRS) from the multi-aminoacyl-tRNA synthetase complex in human macrophage-like differentiated THP-1 cells and its subsequent secretion. The secreted KRS acted to increase the production of proinflammatory cytokines and chemokines. Thus, KRS may be one of the key factors that mediate transduction of inflammatory signals in the STEC-infected host.


Subject(s)
Lysine-tRNA Ligase/metabolism , Macrophages/immunology , Shiga Toxins/pharmacology , Amino Acyl-tRNA Synthetases/isolation & purification , Amino Acyl-tRNA Synthetases/metabolism , Cell Line , Chemokines/metabolism , Cytokines/metabolism , Escherichia coli O157/genetics , Humans , Shiga Toxins/metabolism , Shiga Toxins/toxicity , Shiga-Toxigenic Escherichia coli
12.
J Cell Sci ; 128(15): 2891-902, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26071526

ABSTRACT

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


Subject(s)
Endocytosis/physiology , Protein Transport/physiology , R-SNARE Proteins/metabolism , Shiga Toxin 1/pharmacology , Shiga Toxins/pharmacology , Vesicle-Associated Membrane Protein 2/metabolism , Vesicle-Associated Membrane Protein 3/metabolism , Cell Line , Cell Membrane/physiology , Clathrin/metabolism , ErbB Receptors/metabolism , HeLa Cells , Humans , Protein Binding/genetics , R-SNARE Proteins/genetics , RNA Interference , RNA, Small Interfering , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Shiga Toxins/metabolism , Transferrin/metabolism , Vesicle-Associated Membrane Protein 2/genetics , Vesicle-Associated Membrane Protein 3/genetics
13.
Autophagy ; 11(2): 344-54, 2015.
Article in English | MEDLINE | ID: mdl-25831014

ABSTRACT

Shiga toxins (Stxs) are a family of cytotoxic proteins that lead to the development of bloody diarrhea, hemolytic-uremic syndrome, and central nervous system complications caused by bacteria such as S. dysenteriae, E. coli O157:H7 and E. coli O104:H4. Increasing evidence indicates that macroautophagy (autophagy) is a key factor in the cell death induced by Stxs. However, the associated mechanisms are not yet clear. This study showed that Stx2 induces autophagic cell death in Caco-2 cells, a cultured line model of human enterocytes. Inhibition of autophagy using pharmacological inhibitors, such as 3-methyladenine and bafilomycin A1, or silencing of the autophagy genes ATG12 or BECN1 decreased the Stx2-induced death in Caco-2 cells. Furthermore, there were numerous instances of dilated endoplasmic reticulum (ER) in the Stx2-treated Caco-2 cells, and repression of ER stress due to the depletion of viable candidates of DDIT3 and NUPR1. These processes led to Stx2-induced autophagy and cell death. Finally, the data showed that the pseudokinase TRIB3-mediated DDIT3 expression and AKT1 dephosphorylation upon ER stress were triggered by Stx2. Thus, the data indicate that Stx2 causes autophagic cell death via the ER stress pathway in intestinal epithelial cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Epithelial Cells/drug effects , Shiga Toxins/pharmacology , Animals , Cell Death/drug effects , Cells, Cultured , Epithelial Cells/cytology , Escherichia coli , Humans , Mice, Inbred C57BL , Transcription Factor CHOP
14.
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
15.
Curr Top Microbiol Immunol ; 357: 41-65, 2012.
Article in English | MEDLINE | ID: mdl-22057792

ABSTRACT

Shiga toxins and ricin are potent inhibitors of protein synthesis. In addition to causing inhibition of protein synthesis, these toxins activate proinflammatory signaling cascades that may contribute to the severe diseases associated with toxin exposure. Treatment of cells with Shiga toxins and ricin have been shown to activate a number of signaling pathways including those associated with the ribotoxic stress response, Nuclear factor kappa B activation, inflammasome activation, the unfolded protein response, mTOR signaling, hemostasis, and retrograde trafficking. In this chapter, we review our current understanding of these signaling pathways as they pertain to intoxication by Shiga toxins and ricin.


Subject(s)
Ricin/pharmacology , Shiga Toxins/pharmacology , Signal Transduction/drug effects , Animals , Carrier Proteins/metabolism , Endoplasmic Reticulum Stress , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , NF-kappa B/metabolism , Peptide Chain Initiation, Translational , Protein Transport , Shiga Toxins/metabolism , Stress, Physiological
16.
Mol Cancer Ther ; 10(10): 1918-28, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21788400

ABSTRACT

Pancreatic carcinoma is one of the most aggressive tumor entities, and standard chemotherapy provides only modest benefit. Therefore, specific targeting of pancreatic cancer for early diagnosis and therapeutic intervention is of great interest. We have previously shown that the cellular receptor for Shiga toxin B (STxB), the glycosphingolipid globotriaosylceramide (Gb(3) or CD77) is strongly increased in colorectal adenocarcinoma and their metastases. Here, we report an upregulation of Gb(3) in pancreatic adenocarcinoma (21 of 27 cases) as compared with matched normal tissue (n = 27). The mean expression was highly significantly increased from 30 ± 16 ng Gb(3)/mg tissue in normal pancreas to 61 ± 41 ng Gb(3)/mg tissue (mean ± SD, P = 0.0006), as evidenced by thin layer chromatography. Upregulation of Gb(3) levels did not depend on tumor stage or grading and showed no correlation with clinical outcome. Tumor cells and endothelial cells were identified as the source of increased Gb(3) expression by immunocytochemistry. Pancreatic cancer cell lines showed rapid intracellular uptake of STxB to the Golgi apparatus, following the retrograde pathway. The therapeutic application of STxB was tested by specific delivery of covalently coupled SN38, an active metabolite of the topoisomerase I inhibitor irinotecan. The cytotoxic effect of the STxB-SN38 compound in pancreatic cancer cell lines was increased more than 100-fold compared with irinotecan. Moreover, this effect was effectively blocked by competing incubation with nonlabeled STxB, showing the specificity of the targeting. Thus, STxB constitutes a promising new tool for specific targeting of pancreatic cancer.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/biosynthesis , Camptothecin/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Shiga Toxins/pharmacology , Adult , Aged , Aged, 80 and over , Antigens, Tumor-Associated, Carbohydrate/metabolism , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Cell Line, Tumor , Female , HT29 Cells , Humans , Immunohistochemistry , Irinotecan , Male , Middle Aged , Shiga Toxins/pharmacokinetics , Topoisomerase I Inhibitors/pharmacology
17.
Traffic ; 11(5): 626-36, 2010 May.
Article in English | MEDLINE | ID: mdl-20163571

ABSTRACT

We used multiple approaches to investigate the coordination of trans and medial Rab proteins in the regulation of intra-Golgi retrograde trafficking. We reasoned that medially located Rab33b might act downstream of the trans Golgi Rab, Rab6, in regulating intra-Golgi retrograde trafficking. We found that knockdown of Rab33b, like Rab6, suppressed conserved oligomeric Golgi (COG) complex- or Zeste White 10 (ZW10)-depletion induced disruption of the Golgi ribbon in HeLa cells. Moreover, efficient GTP-restricted Rab6 induced relocation of Golgi enzymes to the endoplasmic reticulum (ER) was Rab33b-dependent, but not vice versa, suggesting that the two Rabs act sequentially in an intra-Golgi Rab cascade. In support of this hypothesis, we found that overexpression of GTP-Rab33b induced the dissociation of Rab6 from Golgi membranes in vivo. In addition, the transport of Shiga-like toxin B fragment (SLTB) from the trans to cis Golgi and ER required Rab33b. Surprisingly, depletion of Rab33b had little, if any, immediate effect on cell growth and multiplication. Furthermore, anterograde trafficking of tsO45G protein through the Golgi apparatus was normal. We suggest that the Rab33b/Rab6 regulated intra-Golgi retrograde trafficking pathway must coexist with other Golgi trafficking pathways. In conclusion, we provide the first evidence that Rab33b and Rab6 act to coordinate a major intra-Golgi retrograde trafficking pathway. This coordination may have parallels with Rab conversion/cascade events that regulate endosome, phagosome and exocytic processes.


Subject(s)
Golgi Apparatus/metabolism , Homeostasis/genetics , Biological Transport/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Golgi Apparatus/drug effects , Golgi Apparatus/genetics , Guanosine Triphosphate/genetics , Guanosine Triphosphate/metabolism , Guanosine Triphosphate/pharmacology , HeLa Cells , Humans , Protein Transport/genetics , Shiga Toxins/genetics , Shiga Toxins/metabolism , Shiga Toxins/pharmacology
18.
Nat Rev Microbiol ; 8(2): 105-16, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20023663

ABSTRACT

Shiga toxin-producing Escherichia coli is an emergent pathogen that can induce haemolytic uraemic syndrome. The toxin has received considerable attention not only from microbiologists but also in the field of cell biology, where it has become a powerful tool to study intracellular trafficking. In this Review, we summarize the Shiga toxin family members and their structures, receptors, trafficking pathways and cellular targets. We discuss how Shiga toxin affects cells not only by inhibiting protein biosynthesis but also through the induction of signalling cascades that lead to apoptosis. Finally, we discuss how Shiga toxins might be exploited in cancer therapy and immunotherapy.


Subject(s)
Neoplasms/therapy , Shiga Toxins , Shiga-Toxigenic Escherichia coli/metabolism , Animals , Antigens, Tumor-Associated, Carbohydrate/metabolism , Apoptosis , Cell Membrane/drug effects , Humans , Immunotherapy , Shiga Toxins/chemistry , Shiga Toxins/metabolism , Shiga Toxins/pharmacology , Shiga Toxins/therapeutic use , Shiga-Toxigenic Escherichia coli/pathogenicity , Signal Transduction
19.
Mol Biol Cell ; 20(20): 4303-12, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19692570

ABSTRACT

Shiga-toxin-producing Escherichia coli remain a food-borne health threat. Shiga toxin is endocytosed by intestinal epithelial cells and transported retrogradely through the secretory pathway. It is ultimately translocated to the cytosol where it inhibits protein translation. We found that Shiga toxin transport through the secretory pathway was dependent on the cytoskeleton. Recent studies reveal that Shiga toxin activates signaling pathways that affect microtubule reassembly and dynein-dependent motility. We propose that Shiga toxin alters cytoskeletal dynamics in a way that facilitates its transport through the secretory pathway. We have now found that Rho GTPases regulate the endocytosis and retrograde motility of Shiga toxin. The expression of RhoA mutants inhibited endocytosis of Shiga toxin. Constitutively active Cdc42 or knockdown of the Cdc42-specific GAP, ARHGAP21, inhibited the transport of Shiga toxin to the juxtanuclear Golgi apparatus. The ability of Shiga toxin to stimulate microtubule-based transferrin transport also required Cdc42 and ARHGAP21 function. Shiga toxin addition greatly decreases the levels of active Cdc42-GTP in an ARHGAP21-dependent manner. We conclude that ARHGAP21 and Cdc42-based signaling regulates the dynein-dependent retrograde transport of Shiga toxin to the Golgi apparatus.


Subject(s)
Cytoskeleton/physiology , GTPase-Activating Proteins/physiology , Shiga Toxins/metabolism , cdc42 GTP-Binding Protein/physiology , Animals , Chlorocebus aethiops , Endocytosis/physiology , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/genetics , Gene Knockdown Techniques , Golgi Apparatus/metabolism , Guanosine Triphosphate/physiology , Microscopy, Confocal , Microtubules/physiology , Protein Transport/physiology , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/physiology , Shiga Toxins/pharmacology , Transferrin/physiology , Vero Cells/metabolism , cdc42 GTP-Binding Protein/antagonists & inhibitors , cdc42 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/physiology , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/physiology
20.
J Bacteriol ; 191(16): 5116-22, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19502393

ABSTRACT

Bacterially derived exotoxins kill eukaryotic cells by inactivating factors and/or pathways that are universally conserved among eukaryotic organisms. The genes that encode these exotoxins are commonly found in bacterial viruses (bacteriophages). In the context of mammals, these toxins cause diseases ranging from cholera to diphtheria to enterohemorrhagic diarrhea. Phage-carried exotoxin genes are widespread in the environment and are found with unexpectedly high frequency in regions lacking the presumed mammalian "targets," suggesting that mammals are not the primary targets of these exotoxins. We suggest that such exotoxins may have evolved for the purpose of bacterial antipredator defense. We show here that Tetrahymena thermophila, a bacterivorous predator, is killed when cocultured with bacteria bearing a Shiga toxin (Stx)-encoding temperate bacteriophage. In cocultures with Tetrahymena, the Stx-encoding bacteria display a growth advantage over those that do not produce Stx. Tetrahymena is also killed by purified Stx. Disruption of the gene encoding the StxB subunit or addition of an excess of the nontoxic StxB subunit substantially reduced Stx holotoxin toxicity, suggesting that this subunit mediates intake and/or trafficking of Stx by Tetrahymena. Bacterially mediated Tetrahymena killing was blocked by mutations that prevented the bacterial SOS response (recA mutations) or by enzymes that breakdown H(2)O(2) (catalase), suggesting that the production of H(2)O(2) by Tetrahymena signals its presence to the bacteria, leading to bacteriophage induction and production of Stx.


Subject(s)
Microbial Viability/genetics , Shiga Toxins/pharmacology , Tetrahymena thermophila/drug effects , Tetrahymena thermophila/microbiology , Animals , Bacteriophages/genetics , Bacteriophages/metabolism , Bacteriophages/physiology , Coculture Techniques , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Hydrogen Peroxide/metabolism , Protein Synthesis Inhibitors/pharmacology , Shiga Toxins/genetics , Shiga Toxins/metabolism , Tetrahymena thermophila/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...