Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Biochem Biophys Res Commun ; 629: 95-100, 2022 11 12.
Article in English | MEDLINE | ID: mdl-36115284

ABSTRACT

Subtilase cytotoxin (SubAB) is a major virulence factor produced by eae-negative Shiga-toxigenic Escherichia coli (STEC) that can cause fatal systemic complications. SubAB binds to target cells through multivalent interactions between its B-subunit pentamer and receptor molecules such as glycoproteins with a terminal N-glycolylneuraminic acid (Neu5Gc). We screened randomized multivalent peptide libraries synthesized on a cellulose membrane and identified a series of tetravalent peptides that efficiently bind to the receptor-binding region of the SubAB B-subunit pentamer. These peptides competitively inhibited the binding of the B-subunit to a receptor-mimic molecule containing clustered Neu5Gc (Neu5Gc-polymer). We selected the peptide with the highest inhibitory efficacy, FFP-tet, and covalently bound it to beads to synthesize FFP-tet-beads, a highly clustered SubAB absorber that displayed potency to absorb SubAB cytotoxicity through direct binding to the toxin. The efficacy of FFP-tet-beads to absorb SubAB cytotoxicity in solution was similar to that of Neu5Gc-polymer, suggesting that FFP-tet-beads might be an effective therapeutic agent against complications arising from eae-negative STEC infection.


Subject(s)
Escherichia coli Proteins , Shiga-Toxigenic Escherichia coli , Carrier Proteins/metabolism , Cellulose/metabolism , Cytotoxins , Escherichia coli Proteins/metabolism , Peptide Library , Polymers/metabolism , Shiga-Toxigenic Escherichia coli/genetics , Shiga-Toxigenic Escherichia coli/metabolism , Subtilisins/toxicity , Virulence Factors/metabolism
2.
Toxins (Basel) ; 13(8)2021 07 29.
Article in English | MEDLINE | ID: mdl-34437406

ABSTRACT

Shiga toxin-producing E. coli (STEC) produces Stx1 and/or Stx2, and Subtilase cytotoxin (SubAB). Since these toxins may be present simultaneously during STEC infections, the purpose of this work was to study the co-action of Stx2 and SubAB. Stx2 + SubAB was assayed in vitro on monocultures and cocultures of human glomerular endothelial cells (HGEC) with a human proximal tubular epithelial cell line (HK-2) and in vivo in mice after weaning. The effects in vitro of both toxins, co-incubated and individually, were similar, showing that Stx2 and SubAB contribute similarly to renal cell damage. However, in vivo, co-injection of toxins lethal doses reduced the survival time of mice by 24 h and mice also suffered a strong decrease in the body weight associated with a lowered food intake. Co-injected mice also exhibited more severe histological renal alterations and a worsening in renal function that was not as evident in mice treated with each toxin separately. Furthermore, co-treatment induced numerous erythrocyte morphological alterations and an increase of free hemoglobin. This work shows, for the first time, the in vivo effects of Stx2 and SubAB acting together and provides valuable information about their contribution to the damage caused in STEC infections.


Subject(s)
Escherichia coli Proteins/toxicity , Hemolytic-Uremic Syndrome/etiology , Shiga Toxin 2/toxicity , Subtilisins/toxicity , Animals , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Endothelial Cells/drug effects , Epithelial Cells/drug effects , Hemolytic-Uremic Syndrome/pathology , Humans , Kidney/drug effects , Kidney/pathology , Kidney Glomerulus/cytology , Kidney Tubules, Proximal/cytology , Male , Mice, Inbred BALB C
3.
BMC Pharmacol Toxicol ; 22(1): 38, 2021 06 26.
Article in English | MEDLINE | ID: mdl-34172094

ABSTRACT

BACKGROUND: Subtilisin QK is a serine protease in the subtilisin family, and is fermented by Bacillus subtilis QK02. The fibrinolytic activity of subtilisin QK was measured by detecting low molecular weight degradation products using a spectrophotometric method developed by Japan Bio Science Laboratory Co., Ltd. Subtilisin QK powder can maintain its fibrinolytic activity for more than 24 months when it is stored at room temperature and protected from light. Our previous results showed that subtlisin QK directly degraded cross-linked fibrins in the fibrin plate assay and effectively inhibited thrombosis in the mouse thrombus model. The aim of this study was to determine the acute toxicity, potential subchronic toxicity, and safety pharmacology of subtilisin QK in Sprague-Dawley (SD) rats. METHODS: In the acute toxicity study, a single oral dose of 100,000 FU/kg was administered to 10 female and 10 male SD rats. In the 28-day subchronic toxicity, 60 female and 60 male SD rats were randomly assigned to four experimental groups (daily oral dose of 0, 2500, 7500 and 25,000 FU/kg). In the safety pharmacology study, 20 female and 20 male SD rats were randomly assigned to four experimental groups (single oral dose of 0, 500, 1500 and 5000 FU/kg). RESULTS: No death occurred and no adverse effects were observed in the acute toxicity study at a dose of 100,000 FU/kg. In the 28-day subchronic toxicity study, several hematological and blood biochemical parameters showed increases or decreases; however, due to the lack of a dose-response relationship, these differences were considered unrelated to treatment. In the safety pharmacology study, no adverse effects were observed on the central nervous of SD rats post-administration up to a dose of 5000 FU/kg subtilisin QK. CONCLUSION: The results showed that oral consumption of subtilisin QK is of low toxicological concern. No adverse effects were observed at doses of 2500, 7500, and 25,000 FU/kg in the 28-day subchronic toxicity, and the no-observed-adverse-effect level (NOAEL) of subtilisin QK was 25,000 FU/kg.


Subject(s)
Fibrinolytic Agents/toxicity , Subtilisins/toxicity , Administration, Oral , Animals , Female , Fibrinolytic Agents/pharmacology , Male , Rats, Sprague-Dawley , Subtilisins/pharmacology , Toxicity Tests, Acute , Toxicity Tests, Subchronic
4.
Biol Pharm Bull ; 44(2): 211-218, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33281147

ABSTRACT

Subtilisin NAT, a Bacillus subtilisin, is widely applied as a functional food and considered to be one of the most exploitable potential oral thrombolytic agents. Subtilisin QK, another Bacillus subtilisin, is a serine protease fermented by Bacillus subtilis 02 and has a better thrombolytic effect. Therefore, subtilisin QK is typically used for evaluating the safety of Bacillus subtilisins. Here, we conduct several good laboratory practice (GLP)-compliant studies in non-rodent animal, i.e., in Beagle dogs, including acute toxicity, subchronic toxicity, and safety pharmacology studies. No adverse effects were evident in the acute and 28-d subchronic toxicity studies at doses up to 40000 FU/kg and 16000 FU/kg/d, respectively. In evaluating the pharmacological safety of up to 2000FU/kg subtilisin QK, we found no significant differences between the electrocardiograms, blood pressures, and respiration of beagle dogs. These findings suggest the safety of Bacillus subtilisin, providing reliable pharmacological and toxicological data for its development and popularization as a functional food and drug.


Subject(s)
Fibrinolytic Agents/toxicity , Subtilisins/toxicity , Animals , Dogs , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Evaluation, Preclinical , Female , Fibrinolytic Agents/administration & dosage , Male , Subtilisins/administration & dosage , Toxicity Tests, Acute , Toxicity Tests, Subchronic
5.
Biochem Biophys Res Commun ; 525(4): 1068-1073, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32184018

ABSTRACT

Subtilase cytotoxin (SubAB) is a member of bacterial AB5 toxin produced by certain enterohemorrhagic E. coli strains which cleaves host chaperone BiP in endoplasmic reticulum (ER), leading to ER stress-mediated cytotoxicity. Previous study suggested that protein disulfide isomerase (PDI), an enzyme which catalyzes the formation and breakage of disulfide bonds in proteins, regulates AB5 toxin such as cholera toxin by unfolding of A subunit, leading to its translocation into cytosol to induce disease. Although SubAB targets ER and has similar A subunit to that of other AB5 toxins, it is unclear whether PDI can modulate the SubAB function. Here we determined the role of PDI on SubAB-induced BiP cleavage, ER stress response and cytotoxicity in HeLa cells. We found that PDI knockdown significantly suppressed SubAB-induced BiP cleavage and eIF2α phosphorylation. The accumulation of SubAB in ER was perturbed upon PDI knockdown. Finally, cell viability assay showed that PDI knockdown and PDI inhibitor canceled the SubAB-induced cytotoxicity. Present results suggested that SubAB, after cellular uptake, translocates into ER and interacts with BiP that might be modulated by PDI. Identification of pivotal role of host proteins on bacterial toxin to elicit its pathogenesis is necessary basis for development of potential chemotherapy and new diagnostic strategy for control of toxin-producing bacterial infections.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Escherichia coli Proteins/toxicity , Protein Disulfide-Isomerases/metabolism , Subtilisins/toxicity , Cell Death/genetics , Cell Survival/drug effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/genetics , Eukaryotic Initiation Factor-2/metabolism , Gene Knockdown Techniques , HeLa Cells , Heat-Shock Proteins/metabolism , Host Microbial Interactions/genetics , Humans , MAP Kinase Kinase 4/metabolism , Phosphorylation , Protein Disulfide-Isomerases/antagonists & inhibitors , Protein Disulfide-Isomerases/genetics , RNA, Small Interfering
6.
Toxins (Basel) ; 11(12)2019 12 03.
Article in English | MEDLINE | ID: mdl-31816894

ABSTRACT

The subtilase cytotoxin (SubAB) of Shiga toxin-producing Escherichia coli (STEC) is a member of the AB5 toxin family. In the current study, we analyzed the formation of active homo- and hetero-complexes of SubAB variants in vitro to characterize the mode of assembly of the subunits. Recombinant SubA1-His, SubB1-His, SubA2-2-His, and SubB2-2-His subunits, and His-tag-free SubA2-2 were separately expressed, purified, and biochemically characterized by circular dichroism (CD) spectroscopy, size-exclusion chromatography (SEC), and analytical ultracentrifugation (aUC). To confirm their biological activity, cytotoxicity assays were performed with HeLa cells. The formation of AB5 complexes was investigated with aUC and isothermal titration calorimetry (ITC). Binding of SubAB2-2-His to HeLa cells was characterized with flow cytometry (FACS). Cytotoxicity experiments revealed that the analyzed recombinant subtilase subunits were biochemically functional and capable of intoxicating HeLa cells. Inhibition of cytotoxicity by Brefeldin A demonstrated that the cleavage is specific. All His-tagged subunits, as well as the non-tagged SubA2-2 subunit, showed the expected secondary structural compositions and oligomerization. Whereas SubAB1-His complexes could be reconstituted in solution, and revealed a Kd value of 3.9 ± 0.8 µmol/L in the lower micromolar range, only transient interactions were observed for the subunits of SubAB2-2-His in solution, which did not result in any binding constant when analyzed with ITC. Additional studies on the binding characteristics of SubAB2-2-His on HeLa cells revealed that the formation of transient complexes improved binding to the target cells. Conclusively, we hypothesize that SubAB variants exhibit different characteristics in their binding behavior to their target cells.


Subject(s)
Escherichia coli Proteins , Protein Subunits , Subtilisins , Cell Survival/drug effects , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/toxicity , HeLa Cells , Humans , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/toxicity , Shiga-Toxigenic Escherichia coli , Subtilisins/chemistry , Subtilisins/genetics , Subtilisins/toxicity
7.
Toxins (Basel) ; 11(11)2019 11 07.
Article in English | MEDLINE | ID: mdl-31703347

ABSTRACT

Hemolytic uremic syndrome (HUS) is a consequence of Shiga toxin (Stx)-producing Escherichia coli (STEC) infection and is the most frequent cause of acute renal failure (ARF) in children. Subtilase cytotoxin (SubAB) has also been associated with HUS pathogenesis. We previously reported that Stx2 and SubAB cause different effects on co-cultures of human renal microvascular endothelial cells (HGEC) and human proximal tubular epithelial cells (HK-2) relative to HGEC and HK-2 monocultures. In this work we have analyzed the secretion of pro-inflammatory cytokines by co-cultures compared to monocultures exposed or not to Stx2, SubAB, and Stx2+SubAB. Under basal conditions, IL-6, IL-8 and TNF-α secretion was different between monocultures and co-cultures. After toxin treatments, high concentrations of Stx2 and SubAB decreased cytokine secretion by HGEC monocultures, but in contrast, low toxin concentrations increased their release. Toxins did not modulate the cytokine secretion by HK-2 monocultures, but increased their release in the HK-2 co-culture compartment. In addition, HK-2 monocultures were stimulated to release IL-8 after incubation with HGEC conditioned media. Finally, Stx2 and SubAB were detected in HGEC and HK-2 cells from the co-cultures. This work describes, for the first time, the inflammatory responses induced by Stx2 and SubAB, in a crosstalk model of renal endothelial and epithelial cells.


Subject(s)
Cytokines/metabolism , Endothelial Cells/drug effects , Epithelial Cells/drug effects , Escherichia coli Proteins/toxicity , Kidney Tubules, Proximal/drug effects , Microvessels/drug effects , Shiga Toxin 2/toxicity , Subtilisins/toxicity , Cell Communication/drug effects , Cell Communication/immunology , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Coculture Techniques , Drug Synergism , Endothelial Cells/immunology , Epithelial Cells/immunology , Hemolytic-Uremic Syndrome , Humans , Kidney/blood supply
8.
Toxins (Basel) ; 9(7)2017 07 18.
Article in English | MEDLINE | ID: mdl-28718802

ABSTRACT

Hemolytic uremic syndrome (HUS) is one of the most common causes of acute renal failure in children. The majority of cases are associated with Shiga toxin (Stx)-producing Escherichia coli (STEC). In Argentina, HUS is endemic and presents the highest incidence rate in the world. STEC strains expressing Stx type 2 (Stx2) are responsible for the most severe cases of this pathology. Subtilase cytotoxin (SubAB) is another STEC virulence factor that may contribute to HUS pathogenesis. To date, neither a licensed vaccine nor effective therapy for HUS is available for humans. Considering that Ouabain (OUA) may prevent the apoptosis process, in this study we evaluated if OUA is able to avoid the damage caused by Stx2 and SubAB on human glomerular endothelial cells (HGEC) and the human proximal tubule epithelial cell (HK-2) line. HGEC and HK-2 were pretreated with OUA and then incubated with the toxins. OUA protected the HGEC viability from Stx2 and SubAB cytotoxic effects, and also prevented the HK-2 viability from Stx2 effects. The protective action of OUA on HGEC and HK-2 was associated with a decrease in apoptosis and an increase in cell proliferation. Our data provide evidence that OUA could be considered as a therapeutic strategy to avoid the renal damage that precedes HUS.


Subject(s)
Escherichia coli Proteins/toxicity , Ouabain/pharmacology , Protective Agents/pharmacology , Shiga Toxin 2/toxicity , Subtilisins/toxicity , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Endothelial Cells/drug effects , Epithelial Cells/drug effects , Humans , Kidney/cytology , Necrosis/chemically induced , Necrosis/prevention & control
9.
Toxicon ; 127: 49-55, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28057514

ABSTRACT

Subtilase cytotoxin (SubAB) is a member of the AB5 cytotoxin family and is produced by certain strains of Shiga toxigenic Escherichia coli. The toxin is known to be lethal to mice, but the pathological mechanisms that contribute to Uremic Hemolytic Syndrome (HUS) are poorly understood. In this study we show that intraperitoneal injection of a sublethal dose of SubAB in rats triggers a systemic response, with ascitic fluid accumulation, heart hypertrophy and damage to the liver, colon and kidney. SubAB treated rats presented microalbuminuria 20 days post inoculation. At this time we found disruption of the glomerular filtration barrier and alteration of the protein reabsorption mechanisms of the proximal tubule. In the kidney, SubAB also triggered an epithelial to mesenchymal transition (Wuyts et al., 1996). These findings indicate that apart from direct cytotoxic effects on renal tissues, SubAB causes significant damage to the other organs, with potential consequences for HUS pathogenesis. IMPORTANCE: Uremic Hemolytic Syndrome is an endemic disease in Argentina, with over 400 hundred new cases each year. We have previously described renal effects of Shiga Toxin and its ability to alter renal protein handling. Bearing in mind that Subtilase Cytotoxin is an emerging pathogenic factor, that it is not routinely searched for in patients with HUS, and that to the date its systemic effects have not been fully clarified we decided to study both its systemic effects, and its renal effects to assess whether SubAB could be contributing to pathology seen in children.


Subject(s)
Escherichia coli Proteins/metabolism , Shiga-Toxigenic Escherichia coli/metabolism , Subtilisins/metabolism , Albuminuria/chemically induced , Animals , Ascites/chemically induced , Cardiomegaly/chemically induced , Colon/drug effects , Colon/pathology , Epithelial-Mesenchymal Transition/drug effects , Escherichia coli Proteins/toxicity , Hemolytic-Uremic Syndrome/etiology , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/pathology , Male , Rats, Sprague-Dawley , Subtilisins/toxicity
10.
PLoS One ; 11(6): e0158180, 2016.
Article in English | MEDLINE | ID: mdl-27336788

ABSTRACT

Postdiarrheal hemolytic uremic syndrome (HUS) affects children under 5 years old and is responsible for the development of acute and chronic renal failure, particularly in Argentina. This pathology is a complication of Shiga toxin (Stx)-producing Escherichia coli infection and renal damage is attributed to Stx types 1 and 2 (Stx1, Stx2) produced by Escherichia coli O157:H7 and many other STEC serotypes. It has been reported the production of Subtilase cytotoxin (SubAB) by non-O157 STEC isolated from cases of childhood diarrhea. Therefore, it is proposed that SubAB may contribute to HUS pathogenesis. The human kidney is the most affected organ because very Stx-sensitive cells express high amounts of biologically active receptor. In this study, we investigated the effects of Stx2 and SubAB on primary cultures of human glomerular endothelial cells (HGEC) and on a human tubular epithelial cell line (HK-2) in monoculture and coculture conditions. We have established the coculture as a human renal proximal tubule model to study water absorption and cytotoxicity in the presence of Stx2 and SubAB. We obtained and characterized cocultures of HGEC and HK-2. Under basal conditions, HGEC monolayers exhibited the lowest electrical resistance (TEER) and the highest water permeability, while the HGEC/HK-2 bilayers showed the highest TEER and the lowest water permeability. In addition, at times as short as 20-30 minutes, Stx2 and SubAB caused the inhibition of water absorption across HK-2 and HGEC monolayers and this effect was not related to a decrease in cell viability. However, toxins did not have inhibitory effects on water movement across HGEC/HK-2 bilayers. After 72 h, Stx2 inhibited the cell viability of HGEC and HK-2 monolayers, but these effects were attenuated in HGEC/HK-2 bilayers. On the other hand, SubAB cytotoxicity shows a tendency to be attenuated by the bilayers. Our data provide evidence about the different effects of these toxins on the bilayers respect to the monolayers. This in vitro model of communication between human renal microvascular endothelial cells and human proximal tubular epithelial cells is a representative model of the human proximal tubule to study the effects of Stx2 and SubAB related to the development of HUS.


Subject(s)
Endothelial Cells/drug effects , Epithelial Cells/drug effects , Escherichia coli Proteins/toxicity , Shiga Toxin 2/toxicity , Subtilisins/toxicity , Biological Transport/drug effects , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Humans , Kidney Glomerulus/cytology , Kidney Glomerulus/drug effects , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects
11.
Food Chem Toxicol ; 88: 87-99, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26740078

ABSTRACT

Subtilisin NAT, commonly known as "nattokinase," is a fibrinolytic enzyme produced by the bacterial strain B. subtilis var. natto, which plays a central role in the fermentation of soybeans into the popular Japanese food natto. Recent studies have reported on the potential anticoagulatory and antihypertensive effects of nattokinase administration in humans, with no indication of adverse effects. To evaluate the safety of nattokinase in a more comprehensive manner, several GLP-compliant studies in rodents and human volunteers have been conducted with the enzyme product, NSK-SD (Japan Bio Science Laboratory Co., Ltd., Japan). Nattokinase was non-mutagenic and non-clastogenic in vitro, and no adverse effects were observed in 28-day and 90-day subchronic toxicity studies conducted in Sprague-Dawley rats at doses up to 167 mg/kg-day and 1000 mg/kg-day, respectively. Mice inoculated with 7.55 × 10(8) CFU of the enzyme-producing bacterial strain showed no signs of toxicity or residual tissue concentrations of viable bacteria. Additionally consumption of 10 mg/kg-day nattokinase for 4 weeks was well tolerated in healthy human volunteers. These findings suggest that the oral consumption of nattokinase is of low toxicological concern. The 90-day oral subchronic NOAEL for nattokinase in male and female Sprague-Dawley rats is 1000 mg/kg-day, the highest dose tested.


Subject(s)
Bacillus subtilis/metabolism , Subtilisins/toxicity , Adult , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Fermentation , Humans , Male , Mice , Middle Aged , Rats , Rats, Sprague-Dawley , Soy Foods , Subtilisins/administration & dosage , Young Adult
12.
PLoS One ; 8(7): e70431, 2013.
Article in English | MEDLINE | ID: mdl-23936204

ABSTRACT

The hemolytic uremic syndrome (HUS) associated with diarrhea is a complication of Shiga toxin (Stx)-producing Escherichia coli (STEC) infection. In Argentina, HUS is endemic and responsible for acute and chronic renal failure in children younger than 5 years old. The human kidney is the most affected organ due to the presence of very Stx-sensitive cells, such as microvascular endothelial cells. Recently, Subtilase cytotoxin (SubAB) was proposed as a new toxin that may contribute to HUS pathogenesis, although its action on human glomerular endothelial cells (HGEC) has not been described yet. In this study, we compared the effects of SubAB with those caused by Stx2 on primary cultures of HGEC isolated from fragments of human pediatric renal cortex. HGEC were characterized as endothelial since they expressed von Willebrand factor (VWF) and platelet/endothelial cell adhesion molecule 1 (PECAM-1). HGEC also expressed the globotriaosylceramide (Gb3) receptor for Stx2. Both, Stx2 and SubAB induced swelling and detachment of HGEC and the consequent decrease in cell viability in a time-dependent manner. Preincubation of HGEC with C-9 -a competitive inhibitor of Gb3 synthesis-protected HGEC from Stx2 but not from SubAB cytotoxic effects. Stx2 increased apoptosis in a time-dependent manner while SubAB increased apoptosis at 4 and 6 h but decreased at 24 h. The apoptosis induced by SubAB relative to Stx2 was higher at 4 and 6 h, but lower at 24 h. Furthermore, necrosis caused by Stx2 was significantly higher than that induced by SubAB at all the time points evaluated. Our data provide evidence for the first time how SubAB could cooperate with the development of endothelial damage characteristic of HUS pathogenesis.


Subject(s)
Endothelial Cells/drug effects , Escherichia coli Proteins/pharmacology , Kidney Glomerulus/drug effects , Shiga Toxin 2/pharmacology , Subtilisins/pharmacology , Antigens, Tumor-Associated, Carbohydrate/metabolism , Apoptosis/drug effects , Cell Survival/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Escherichia coli Proteins/toxicity , Humans , Kidney Glomerulus/metabolism , Necrosis/drug therapy , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Shiga Toxin 2/toxicity , Subtilisins/toxicity , von Willebrand Factor/metabolism
13.
Nihon Saikingaku Zasshi ; 68(3): 299-311, 2013.
Article in Japanese | MEDLINE | ID: mdl-23985936

ABSTRACT

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


Subject(s)
Cholera Toxin/toxicity , Enterohemorrhagic Escherichia coli/metabolism , Escherichia coli Proteins/toxicity , Shiga Toxins/toxicity , Subtilisins/toxicity , Vibrio cholerae/metabolism , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cholera Toxin/antagonists & inhibitors , Cholera Toxin/biosynthesis , Cholera Toxin/chemistry , Disease Outbreaks , Endoplasmic Reticulum Stress/drug effects , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/chemistry , Foodborne Diseases/epidemiology , Foodborne Diseases/microbiology , Humans , Macrophages/immunology , Phagocytosis/drug effects , Protein Structure, Tertiary , Shiga Toxins/antagonists & inhibitors , Shiga Toxins/biosynthesis , Shiga Toxins/chemistry , Subtilisins/antagonists & inhibitors , Subtilisins/biosynthesis , Subtilisins/chemistry , Vaccines, Attenuated , Vibrio cholerae/pathogenicity
14.
Biochem Biophys Res Commun ; 410(4): 852-8, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21703246

ABSTRACT

Subtilase cytotoxin (SubAB) is the prototype of a distinct AB(5) toxin family produced by Shiga toxigenic Escherichia coli. Recent reports disclosed pro-apoptotic pathways triggered by SubAB, whereas its anti-apoptotic signals have not been elucidated. In the present study, we investigated pro-survival signaling elicited by SubAB, especially focusing on extracellular signal-regulated kinase (ERK) and Akt. We found that SubAB activated ERK and Akt, and inhibition of individual kinases enhanced SubAB-triggered apoptosis. SubAB induced endoplasmic reticulum (ER) stress, and other ER stress inducers mimicked the stimulatory effects of SubAB on ERK and Akt. Attenuation of ER stress reduced SubAB-induced phosphorylation of these kinases, suggesting involvement of the unfolded protein response (UPR). SubAB induced activation of protein kinase-like ER kinase (PERK) and phosphorylation of eukaryotic translation initiation factor 2α (eIF2α), and phosphorylation of eIF2α by salubrinal caused activation of ERK and Akt, leading to cell survival. Dominant-negative inhibition of PERK enhanced SubAB-induced apoptosis and reduced phosphorylation of ERK and Akt. Furthermore, the anti-apoptotic effect of eIF2α was significantly reversed by inhibition of ERK and Akt. These results suggest cytoprotective roles of ERK and Akt in SubAB-triggered, ER stress-mediated apoptosis.


Subject(s)
Apoptosis , Endoplasmic Reticulum/drug effects , Escherichia coli Proteins/toxicity , Extracellular Signal-Regulated MAP Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stress, Physiological , Subtilisins/toxicity , Animals , Cell Line , Endoplasmic Reticulum/enzymology , Enzyme Activation , Eukaryotic Initiation Factor-2/metabolism , Rats , eIF-2 Kinase/metabolism
15.
Infect Immun ; 79(4): 1671-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21282417

ABSTRACT

Subtilase cytotoxin (SubAB) is the prototype of a new family of AB(5) cytotoxins produced by Shiga-toxigenic Escherichia coli. Its cytotoxicity is due to its capacity to enter cells and specifically cleave the essential endoplasmic reticulum chaperone BiP. Previous studies have shown that intraperitoneal injection of mice with purified SubAB causes a pathology that overlaps with that seen in human cases of hemolytic-uremic syndrome, as well as dramatic splenic atrophy, suggesting that leukocytes are targeted. Here we investigated SubAB-induced leukocyte changes in the peritoneal cavity, blood, and spleen. After intraperitoneal injection, SubAB bound peritoneal leukocytes (including T and B lymphocytes, neutrophils, and macrophages). SubAB elicited marked leukocytosis, which peaked at 24 h, and increased neutrophil activation in the blood and peritoneal cavity. It also induced a marked redistribution of leukocytes among the three compartments: increases in leukocyte subpopulations in the blood and peritoneal cavity coincided with a significant decline in splenic cells. SubAB treatment also elicited significant increases in the apoptosis rates of CD4(+) T cells, B lymphocytes, and macrophages. These findings indicate that apart from direct cytotoxic effects, SubAB interacts with cellular components of both the innate and the adaptive arm of the immune system, with potential consequences for disease pathogenesis.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli Proteins/toxicity , Leukocytes/metabolism , Leukocytes/pathology , Subtilisins/metabolism , Subtilisins/toxicity , Animals , Apoptosis/immunology , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique , Male , Mice , Mice, Inbred BALB C
16.
J Infect Dis ; 202(9): 1415-23, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20874089

ABSTRACT

Subtilase cytotoxin (SubAB), produced by certain virulent Shiga toxigenic Escherichia coli strains, causes hemolytic uremic syndrome-like pathology in mice, including extensive microvascular thrombosis. SubAB acts by specifically cleaving the essential endoplasmic reticulum chaperone binding immunoglobulin protein (BiP). BiP has been reported to inhibit the activation of tissue factor (TF), the major initiator of extrinsic coagulation. We hypothesized that the apparent prothrombotic effect of SubAB in vivo may involve the stimulation of TF­dependent procoagulant activity. TF­dependent procoagulant activity, TF messenger RNA (mRNA) levels, and BiP cleavage were therefore examined in human macrophage cells and primary human umbilical vein endothelial cells exposed to SubAB. In both types of cells, SubAB significantly increased TF­dependent procoagulant activity, induced TF mRNA expression, and mediated BiP cleavage. No effects were seen when cells were treated with a nonproteolytic mutant toxin, SubAA272B. Our results suggest that the procoagulant effect of SubAB may be dependent on both the up­regulation of TF expression and the activation of TF by means of BiP cleavage.


Subject(s)
Escherichia coli Proteins/toxicity , Heat-Shock Proteins/metabolism , Shiga-Toxigenic Escherichia coli/pathogenicity , Subtilisins/toxicity , Thromboplastin/biosynthesis , Animals , Blood Coagulation/drug effects , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Endothelial Cells/drug effects , Escherichia coli Proteins/metabolism , Gene Expression Profiling , Humans , Macrophages/drug effects , Mice , Subtilisins/metabolism , Thrombosis/chemically induced
17.
Infect Immun ; 78(11): 4691-6, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20713620

ABSTRACT

Subtilase cytotoxin (SubAB) was first isolated from a Shiga toxigenic Escherichia coli (STEC) strain that was responsible for an outbreak of hemolytic-uremic syndrome and is the prototype of a new family of AB(5) cytotoxins. SubAB is a subtilase-like serine protease, and upon uptake by host cells, it is trafficked to the endoplasmic reticulum (ER), where it cleaves the essential ER chaperone BiP (GRP78) with high specificity. Previous work has shown that BiP cleavage by SubAB initiates ER stress-signaling pathways in host cells that eventuate in cell death associated with DNA fragmentation, a hallmark of apoptosis. The present study has investigated the role of the Bcl-2 protein family, which has been shown to regulate ER stress-induced apoptosis in other model systems. Examination of the cytotoxicity of SubAB for wild-type and bax(-/-)/bak(-/-) mouse embryonic fibroblasts and comparison of apoptotic markers in these cells revealed that SubAB cytotoxicity can be predominantly attributed to the activation of apoptotic pathways activated by Bax/Bak. The results of the present study further our understanding of the molecular mechanism whereby SubAB kills eukaryotic cells and contributes to STEC pathogenesis, in addition to consolidating the roles of Bcl-2 family members in the regulation of ER stress-induced apoptosis.


Subject(s)
Apoptosis , Escherichia coli Proteins/toxicity , Gene Expression Regulation , Shiga-Toxigenic Escherichia coli/pathogenicity , Subtilisins/toxicity , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Chlorocebus aethiops , DNA Fragmentation , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Chaperone BiP , Fibroblasts , Mice , Vero Cells
18.
Microb Pathog ; 49(4): 153-63, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20561923

ABSTRACT

Subtilase cytotoxin (SubAB) is an AB(5) cytotoxin produced by some strains of Shiga-toxigenic Escherichia coli. The A subunit is a subtilase-like serine protease and cleaves an endoplasmic reticulum (ER) chaperone, BiP, leading to transient inhibition of protein synthesis and cell cycle arrest at G(1) phase, and inducing caspase-dependent apoptosis via mitochondrial membrane damage in Vero cells. Here we investigated the mechanism of mitochondrial permeabilization in HeLa cells. SubAB-induced cytochrome c release into cytosol did not depend on mitochondrial permeability transition pore (PTP), since cyclosporine A did not suppress cytochrome c release. SubAB did not change the expression of anti-apoptotic Bcl-2 or Bcl-XL and pro-apoptotic Bax or Bak, but triggered Bax and Bak conformational changes and association of Bax with Bak. Silencing using siRNA of both bax and bak genes, but not bax, bak, or bim alone, resulted in reduction of cytochrome c release, caspase-3 activation, DNA ladder formation and cytotoxicity, indicating that Bax and Bak were involved in apoptosis. SubAB activated ER transmembrane transducers, Ire1alpha, and cJun N-terminal kinase (JNK), and induced C/EBF-homologue protein (CHOP). To investigate whether these signals were involved in cytochrome c release by Bax activation, we silenced ire1alpha, jnk or chop; however, silencing did not decrease SubAB-induced cytochrome c release, suggesting that these signals were not necessary for SubAB-induced mitochondrial permeabilization by Bax activation.


Subject(s)
Apoptosis , Escherichia coli Proteins/toxicity , Escherichia coli/pathogenicity , Mitochondrial Membranes/drug effects , Permeability/drug effects , Subtilisins/toxicity , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Caspase 3/metabolism , Cytochromes c/metabolism , Cytoplasm/chemistry , Endoribonucleases/metabolism , Gene Silencing , HeLa Cells , Humans , MAP Kinase Kinase 4/metabolism , Protein Binding , Protein Conformation/drug effects , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/metabolism , Signal Transduction , Transcription Factor CHOP/metabolism , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/chemistry
19.
Kidney Int ; 76(2): 140-4, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19387473

ABSTRACT

Hemolytic-uremic syndrome (HUS) is a systemic disease characterized by microvascular endothelial damage, mainly in the gastrointestinal tract and the kidneys. A major cause of HUS is Shiga toxigenic Escherichia coli (STEC) infection. In addition to Shiga toxin, additional STEC virulence factors may contribute to HUS. One is the newly discovered subtilase cytotoxin (SubAB), which is highly toxic to eukaryotic cells, and when injected intraperitoneally into mice causes pathology resembling that associated with human HUS. Recent data show that SubAB exhibits a strong preference for glycans terminating in alpha2-3-linked N-glycolylneuraminic acid (Neu5Gc), a sialic acid that humans are unable to synthesize, because we genetically lack the necessary enzyme. However, Neu5Gc can still be found on human cells due to metabolic incorporation from the diet. Dietary incorporation happens to be highest in human endothelium and to a lesser extent in the intestinal epithelium, the two affected cell types in STEC-induced HUS. Mammalian-derived foods such as red meat and dairy products appear to be the primary source of dietary Neu5Gc. Ironically, these are also common sources of STEC contamination. Taken together, these findings suggest a 'two-hit' process in the pathogenesis of human SubAB-induced disease. First, humans eat Neu5Gc-rich food, leading to incorporation of Neu5Gc on the surfaces of endothelial and intestinal cells. Second, when exposed to a SubAB-producing STEC strain, the toxin produced would be able to bind to the intestinal epithelial cells, perhaps causing acute gastrointestinal symptoms, and eventually damaging endothelial cells in other organs like the kidney, thereby causing HUS.


Subject(s)
Diet/adverse effects , Escherichia coli Proteins/metabolism , Hemolytic-Uremic Syndrome/etiology , Neuraminic Acids/adverse effects , Subtilisins/metabolism , Animals , Escherichia coli Proteins/toxicity , Hemolytic-Uremic Syndrome/chemically induced , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/pathology , Humans , Meat/adverse effects , Meat/microbiology , Neuraminic Acids/pharmacokinetics , Shiga-Toxigenic Escherichia coli/pathogenicity , Subtilisins/toxicity
20.
Infect Immun ; 77(7): 2919-24, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19380466

ABSTRACT

Subtilase cytotoxin (SubAB) is an AB(5) cytotoxin produced by some strains of Shiga-toxigenic Escherichia coli. The A subunit is a subtilase-like serine protease and cleaves an endoplasmic reticulum chaperone, BiP, leading to transient inhibition of protein synthesis and cell cycle arrest at G(1) phase. Here we show that SubAB, but not the catalytically inactive mutant SubAB(S272A), induced apoptosis in Vero cells, as detected by DNA fragmentation and annexin V binding. SubAB induced activation of caspase-3, -7, and -8. Caspase-3 appeared earlier than caspase-8, and by use of specific caspase inhibitors, it was determined that caspase-3 may be upstream of caspase-8. A general caspase inhibitor blocked SubAB-induced apoptosis, detected by annexin V binding. SubAB also stimulated cytochrome c release from mitochondria, which was not suppressed by caspase inhibitors. In HeLa cells, Apaf-1 small interfering RNA inhibited caspase-3 activation, suggesting that cytochrome c might form an apoptosome, leading to activation of caspase-3. These data suggested that SubAB induced caspase-dependent apoptosis in Vero cells through mitochondrial membrane damage.


Subject(s)
Apoptosis , Escherichia coli Proteins/toxicity , Mitochondrial Membranes/drug effects , Shiga-Toxigenic Escherichia coli/pathogenicity , Subtilisins/toxicity , Animals , Annexin A5/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Caspase 8/metabolism , Chlorocebus aethiops , Cytochromes c/metabolism , DNA Fragmentation , HeLa Cells , Humans , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...