Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Toxins (Basel) ; 15(12)2023 12 11.
Article in English | MEDLINE | ID: mdl-38133199

ABSTRACT

Clostridium perfringens iota-toxin is composed of two separate proteins: a binding protein (Ib) that recognizes a host cell receptor and promotes the cellular uptake of a catalytic protein and (Ia) possessing ADP-ribosyltransferase activity that induces actin cytoskeleton disorganization. Ib exhibits the overall structure of bacterial pore-forming toxins (PFTs). Lipolysis-stimulated lipoprotein receptor (LSR) is defined as a host cell receptor for Ib. The binding of Ib to LSR causes an oligomer formation of Ib in lipid rafts of plasma membranes, mediating the entry of Ia into the cytoplasm. Ia induces actin cytoskeleton disruption via the ADP-ribosylation of G-actin and causes cell rounding and death. The binding protein alone disrupts the cell membrane and induces cytotoxicity in sensitive cells. Host cells permeabilized by the pore formation of Ib are repaired by a Ca2+-dependent plasma repair pathway. This review shows that the cellular uptake of iota-toxin utilizes a pathway of plasma membrane repair and that Ib alone induces cytotoxicity.


Subject(s)
Actins , Clostridium perfringens , Animals , Chlorocebus aethiops , Clostridium perfringens/metabolism , Biological Transport , Actins/metabolism , Vero Cells , ADP Ribose Transferases/chemistry
2.
Front Cell Infect Microbiol ; 12: 824547, 2022.
Article in English | MEDLINE | ID: mdl-35273923

ABSTRACT

Aeromonas sobria is a Gram-negative pathogen that causes food-borne illness. In immunocompromised patients and the elderly, A. sobria opportunistically leads to severe extraintestinal diseases including sepsis, peritonitis, and meningitis. If A. sobria that infects the intestinal tract causes such an extraintestinal infection, the pathogen must pass through the intestinal epithelial barrier. In our earlier study using intestinal cultured cells (T84 cells), we observed that an A. sobria strain with higher A. sobria serine protease (ASP) production caused a marked level of bacterial translocation across the T84 intestinal epithelial monolayer. Herein, we investigated the effect of ASP on tight junctions (TJs) in T84 cells. We observed that ASP acts on TJs and causes the destruction of ZO-1, ZO-2, ZO-3, and claudin-7 (i.e., some of the protein components constituting TJs), especially in the strains with high ASP productivity. Based on the present results together with those of our earlier study, we propose that ASP may cause a disruption of the barrier function of the intestinal epithelium as a whole due to the destruction of TJs (in addition to the destruction of adherens junctions) and that ASP may assist invasion of the pathogens from the intestinal epithelium into deep sites in the human body.


Subject(s)
Aeromonas , Bacterial Translocation , Serine Proteases , Tight Junctions , Aeromonas/enzymology , Cell Line , Humans , Intestinal Mucosa/microbiology , Serine Proteases/metabolism , Tight Junctions/metabolism
3.
Access Microbiol ; 4(2): 000319, 2022.
Article in English | MEDLINE | ID: mdl-35355871

ABSTRACT

Introduction: Methicillin-resistant Staphylococcus aureus (MRSA) is one of the major pathogens of nosocomial infections throughout the world. In the medical field, it is extremely important to this pathogen's trends when considering infection control. Hypothesis/Gap Statement: We hypothesized that clarifying the characteristics of clinically isolated MRSA would contribute to infection control and proper use of antimicrobial agents against MRSA. Aim: The purpose of this study is to elucidate the genetic and biological characteristics of the MRSA isolates found at our hospital and to reveal changes in the spread of this pathogen in the local area where we live. Methodology: Pulse-field gel electrophoresis (PFGE) and polymerase chain reaction were used for the genetic analyses of MRSA isolates. Toxin production by each isolate was examined using toxin-specific detection systems. Results: During the 3 years from 2017 through 2019, over 1000 MRSA strains were isolated at our hospital. Genomic analysis of 237 of these clinical isolates by PFGE revealed 12 PFGE types (types A to L), each consisting of five or more MRSA clinical strains with over 80% genetic similarity. Examination of the SCCmec genotypes found that 219 of 237 isolated MRSA strains (approximately 92%) were SCCmec genotype II or IV and that only four of the isolates carried the Panton-Valentine leukocidin (PVL) gene. Examination of the toxin production of the isolates using staphylococcal enterotoxin detection kits found that most isolates carrying the SCCmec genotype II produced enterotoxin B and/or C, and that most isolates carrying the SCCmec genotype IV produced enterotoxin A. Conclusion: The present results revealed that MRSA isolates with common properties were isolated at certain rates throughout the 3 year study period, suggesting that relatively specific MRSA clones may have settled in the local area around our hospital. We also examine the relationship between antimicrobial usage over time and changes in MRSA isolation rates.

4.
Toxins (Basel) ; 12(5)2020 04 30.
Article in English | MEDLINE | ID: mdl-32365779

ABSTRACT

Epsilon-toxin produced by Clostridium perfringens significantly contributes to the pathogeneses of enterotoxemia in ruminants and multiple sclerosis in humans. Epsilon-toxin forms a heptameric oligomer in the host cell membrane, promoting cell disruption. Here, we investigate the effect of epsilon-toxin on epithelial barrier functions. Epsilon-toxin impairs the barrier integrity of Madin-Darby Canine Kidney (MDCK) cells, as demonstrated by decreased transepithelial electrical resistance (TEER), increased paracellular flux marker permeability, and the decreased cellular localization of junctional proteins, such as occludin, ZO-1, and claudin-1. U73122, an endogenous phospholipase C (PLC) inhibitor, inhibited the decrease in TEER and the increase in the permeability of flux marker induced by epsilon-toxin. The application of epsilon-toxin to MDCK cells resulted in the biphasic formation of 1,2-diacylglycerol (DAG) and inositol-1,4,5-triphosphate (IP3). U73122 blocked the formation of DAG and IP3 induced by the toxin. Epsilon-toxin also specifically activated endogenous PLC-γ1. Epsilon-toxin dose-dependently increased the cytosolic calcium ion concentration ([Ca2+]i). The toxin-induced elevation of [Ca2+]i was inhibited by U73122. Cofilin is a key regulator of actin cytoskeleton turnover and tight-junction (TJ) permeability regulation. Epsilon-toxin caused cofilin dephosphorylation. These results demonstrate that epsilon-toxin induces Ca2+ influx through activating the phosphorylation of PLC-γ1 and then causes TJ opening accompanied by cofilin dephosphorylation.


Subject(s)
Bacterial Toxins/toxicity , Calcium Signaling/drug effects , Calcium/metabolism , Epithelial Cells/drug effects , Tight Junctions/drug effects , Actin Depolymerizing Factors/metabolism , Animals , Dogs , Electric Impedance , Epithelial Cells/metabolism , Epithelial Cells/pathology , Madin Darby Canine Kidney Cells , Permeability , Phospholipase C gamma/metabolism , Phosphorylation , Tight Junctions/metabolism , Tight Junctions/pathology
5.
Front Microbiol ; 11: 613650, 2020.
Article in English | MEDLINE | ID: mdl-33488556

ABSTRACT

Aeromonas spp. are Gram-negative rod-shaped bacteria ubiquitously distributed in diverse water sources. Several Aeromonas spp. are known as human and fish pathogens. Recently, attention has been focused on the relationship between bacterial biofilm formation and pathogenicity or drug resistance. However, there have been few reports on biofilm formation by Aeromonas. This study is the first to examine the in vitro formation and components of the biofilm of several Aeromonas clinical and environmental strains. A biofilm formation assay using 1% crystal violet on a polystyrene plate revealed that most Aeromonas strains used in this study formed biofilms but one strain did not. Analysis of the basic components contained in the biofilms formed by Aeromonas strains confirmed that they contained polysaccharides containing GlcNAc, extracellular nucleic acids, and proteins, as previously reported for the biofilms of other bacterial species. Among these components, we focused on several proteins fractionated by SDS-PAGE and determined their amino acid sequences. The results showed that some proteins existing in the Aeromonas biofilms have amino acid sequences homologous to functional proteins present in the outer membrane of Gram-negative bacteria. This result suggests that outer membrane components may affect the biofilm formation of Aeromonas strains. It is known that Gram-negative bacteria often release extracellular membrane vesicles from the outer membrane, so we think that the outer membrane-derived proteins found in the Aeromonas biofilms may be derived from such membrane vesicles. To examine this idea, we next investigated the ability of Aeromonas strains to form outer membrane vesicles (OMVs). Electron microscopic analysis revealed that most Aeromonas strains released OMVs outside the cells. Finally, we purified OMVs from several Aeromonas strains and examined their effect on the biofilm formation. We found that the addition of OMVs dose-dependently promoted biofilm formation, except for one strain that did not form biofilms. These results suggest that the OMVs released from the bacterial cells are closely related to the biofilm formation of Aeromonas strains.

6.
PLoS One ; 14(8): e0221344, 2019.
Article in English | MEDLINE | ID: mdl-31419250

ABSTRACT

Aeromonas sobria is a pathogen causing food-borne illness. In immunocompromised patients and the elderly, A. sobria can leave the intestinal tract, and this opportunistically leads to severe extraintestinal diseases including sepsis, peritonitis, and meningitis. To cause such extraintestinal diseases, A. sobria must pass through the intestinal epithelial barrier. The mechanism of such bacterial translocation has not been established. Herein we used intestinal (T84) cultured cells to investigate the effect of A. sobria serine protease (ASP) on junctional complexes that maintain the intercellular adhesion of the intestinal epithelium. When several A. sobria strains were inoculated into T84 monolayer grown on Transwell inserts, the strain with higher ASP production largely decreased the value of transepithelial electrical resistance exhibited by the T84 monolayer and markedly caused bacterial translocation from the apical surface into the basolateral side of T84 monolayer. Further experiments revealed that ASP acts on adherens junctions (AJs) and causes the destruction of both nectin-2 and afadin, which are protein components constituting AJs. Other studies have not revealed the bacterial pathogenic factors that cause the destruction of both nectin-2 and afadin, and our present results thus provide the first report that the bacterial extracellular protease ASP affects these molecules. We speculate that the destruction of nectin-2 and afadin by the action of ASP increases the ability of A. sobria to pass through intestinal epithelial tissue and contributes to the severity of pathological conditions.


Subject(s)
Aeromonas/pathogenicity , Bacterial Proteins/metabolism , Foodborne Diseases/pathology , Intestinal Mucosa/pathology , Serine Proteases/metabolism , Aeromonas/metabolism , Bacterial Translocation , Cell Culture Techniques , Cell Line , Foodborne Diseases/microbiology , Humans , Intestinal Mucosa/cytology , Kinesins/metabolism , Myosins/metabolism , Nectins/metabolism
7.
Toxins (Basel) ; 11(4)2019 04 22.
Article in English | MEDLINE | ID: mdl-31013617

ABSTRACT

Clostridium perfringens strains B and C cause fatal intestinal diseases in animals. The secreted pore-forming toxin delta-toxin is one of the virulence factors of the strains, but the mechanism of intestinal pathogenesis is unclear. Here, we investigated the effects of delta-toxin on the mouse ileal loop. Delta-toxin caused fluid accumulation and intestinal permeability to fluorescein isothiocyanate (FITC)-dextran in the mouse ileal loop in a dose- and time-dependent manner. Treatment with delta-toxin induced significant histological damage and shortening of villi. Delta-toxin activates a disintegrin and metalloprotease (ADAM) 10, leading to the cleavage of E-cadherin, the epithelial adherens junction protein, in human intestinal epithelial Caco-2 cells. In this study, E-cadherin immunostaining in mouse intestinal epithelial cells was almost undetectable 1 h after toxin treatment. ADAM10 inhibitor (GI254023X) blocked the toxin-induced fluid accumulation and E-cadherin loss in the mouse ileal loop. Delta-toxin stimulated the shedding of intestinal epithelial cells. The shedding cells showed the accumulation of E-cadherin in intracellular vesicles and the increased expression of active caspase-3. Our findings demonstrate that delta-toxin causes intestinal epithelial cell damage through the loss of E-cadherin cleaved by ADAM10.


Subject(s)
Bacterial Toxins/toxicity , Intestine, Small/drug effects , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Cadherins/metabolism , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Membrane Proteins/metabolism , Mice, Inbred ICR
8.
Commun Biol ; 2: 45, 2019.
Article in English | MEDLINE | ID: mdl-30729183

ABSTRACT

During bacterial infection, granulocyte colony-stimulating factor (G-CSF) is produced and accelerates neutrophil production from their progenitors. This process, termed granulopoiesis, strengthens host defense, but Clostridium perfringens α-toxin impairs granulopoiesis via an unknown mechanism. Here, we tested whether G-CSF accounts for the α-toxin-mediated impairment of granulopoiesis. We find that α-toxin dramatically accelerates G-CSF production from endothelial cells in response to Toll-like receptor 2 (TLR2) agonists through activation of the c-Jun N-terminal kinase (JNK) signaling pathway. Meanwhile, α-toxin inhibits G-CSF-mediated cell proliferation of Ly-6G+ neutrophils by inducing degradation of G-CSF receptor (G-CSFR). During sepsis, administration of α-toxin promotes lethality and tissue injury accompanied by accelerated production of inflammatory cytokines in a TLR4-dependent manner. Together, our results illustrate that α-toxin disturbs G-CSF-mediated granulopoiesis by reducing the expression of G-CSFR on neutrophils while augmenting septic shock due to excess inflammatory cytokine release, which provides a new mechanism to explain how pathogenic bacteria modulate the host immune system.


Subject(s)
Bacterial Toxins/toxicity , Calcium-Binding Proteins/toxicity , Clostridium perfringens/pathogenicity , Gas Gangrene/genetics , Granulocyte Colony-Stimulating Factor/genetics , Lipopolysaccharides/toxicity , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Shock, Septic/genetics , Type C Phospholipases/toxicity , Animals , Clostridium perfringens/genetics , Clostridium perfringens/immunology , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Female , Gas Gangrene/immunology , Gas Gangrene/microbiology , Gas Gangrene/mortality , Gene Expression Regulation , Granulocyte Colony-Stimulating Factor/immunology , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoiesis/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/microbiology , Receptors, Granulocyte Colony-Stimulating Factor/immunology , Shock, Septic/immunology , Shock, Septic/microbiology , Shock, Septic/mortality , Signal Transduction , Survival Analysis , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
9.
Biochim Biophys Acta Biomembr ; 1860(2): 428-433, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28988777

ABSTRACT

Clostridium perfringens delta-toxin is a ß-barrel-pore-forming toxin (ß-PFT) and a presumptive virulence factor of type B and C strains, which are causative organisms of fatal intestinal diseases in animals. We showed previously that delta-toxin causes cytotoxicity via necrosis in sensitive cells. Here, we examined the effect of delta-toxin on intestinal membrane integrity. Delta-toxin led to a reduction in transepithelial electrical resistance (TEER) and increased the permeability of fluorescence isothiocyanate-conjugated dextran in human intestinal epithelial Caco-2 cells without changing the tight junction proteins, such as zonula occludens-1 (ZO-1), occludin, and claudin-1. On the other hand, delta-toxin reduced the cellular levels of adherence junction protein E-cadherin before cell injury. A disintegrin and metalloprotease (ADAM) 10 facilitates E-cadherin cleavage and was identified as the cellular receptor for alpha-toxin, a ß-PFT produced by Staphylococcus aureus. ADAM10 inhibitor (GI254023X) blocked the toxin-induced decrease in TEER and cleavage of E-cadherin. Delta-toxin enhanced ADAM10 activity in a dose- and time-dependent manner. Furthermore, delta-toxin colocalized with ADAM10. These results indicated that ADAM10 plays a key role in delta-toxin-induced intestinal injury.


Subject(s)
Bacterial Toxins/pharmacology , Cell Membrane Permeability/drug effects , Epithelial Cells/drug effects , Intestinal Mucosa/drug effects , ADAM10 Protein/metabolism , Caco-2 Cells , Cadherins/metabolism , Claudin-1/metabolism , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Humans , Immunoblotting , Intestinal Mucosa/metabolism , Occludin/metabolism , Time Factors , Zonula Occludens-1 Protein/metabolism
10.
PLoS One ; 12(10): e0186392, 2017.
Article in English | MEDLINE | ID: mdl-29023605

ABSTRACT

Aeromonas sobria serine protease (ASP) is an extracellular serine protease secreted by the organism. Here, we identified the amino acid residue of ASP that contributes to substrate specificity by using both synthetic peptides and biological protein components. The results showed that the arginine residue at position 566 (Arg-566) of ASP, which is located in the extra occluding region of ASP close to an entrance of the catalytic cavity, is involved in the substrate specificity. A substitutional point mutation of the Arg-566 residue of ASP to Ala residue (ASP[R566A]) caused a decrease of the proteolytic efficiency for a certain substrate. In addition, ASP lost the ability to recognize the primary substrate by such a point mutation, and ASP[R566A] reacted to a wide range of synthetic substrates. It is likely that Arg-566 causes an interaction with the amino acid residue at position P3 of the substrate, which is the third amino acid residue upstream from the cleavage site. Another study using ORF2 protein, a chaperone protein of ASP, further suggested that Arg-566 could also play an important role in interaction with ORF2. We therefore conclude that the Arg-566 residue of ASP is likely responsible for the selection of substrates.


Subject(s)
Aeromonas/enzymology , Arginine/metabolism , Bacterial Proteins/metabolism , Serine Proteases/metabolism , Amino Acid Sequence , Arginine/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Fibrinogen/metabolism , Humans , Kininogens/metabolism , Molecular Chaperones/metabolism , Mutagenesis, Site-Directed , Proteolysis , Serine Proteases/chemistry , Serine Proteases/genetics , Substrate Specificity
11.
Toxins (Basel) ; 9(8)2017 08 11.
Article in English | MEDLINE | ID: mdl-28800062

ABSTRACT

Clostridium perfringens iota-toxin and Clostridium botulinum C2 toxin are composed of two non-linked proteins, one being the enzymatic component and the other being the binding/translocation component. These latter components recognize specific receptors and oligomerize in plasma membrane lipid-rafts, mediating the uptake of the enzymatic component into the cytosol. Enzymatic components induce actin cytoskeleton disorganization through the ADP-ribosylation of actin and are responsible for cell rounding and death. This review focuses upon the recent advances in cellular internalization of clostridial binary toxins.


Subject(s)
ADP Ribose Transferases/chemistry , Bacterial Toxins/chemistry , Botulinum Toxins/chemistry , Clostridium perfringens/metabolism , Actins/metabolism , Animals , Biological Transport , Chlorocebus aethiops , Humans , Vero Cells
12.
Sci Rep ; 7(1): 5217, 2017 07 12.
Article in English | MEDLINE | ID: mdl-28701754

ABSTRACT

Clostridium perfringens α-toxin induces hemolysis of erythrocytes from various species, but it has not been elucidated whether the toxin affects erythropoiesis. In this study, we treated bone marrow cells (BMCs) from mice with purified α-toxin and found that TER119+ erythroblasts were greatly decreased by the treatment. A variant α-toxin defective in enzymatic activities, phospholipase C and sphingomyelinase, had no effect on the population of erythroblasts, demonstrating that the decrease in erythroblasts was dependent of its enzymatic activities. α-Toxin reduced the CD71+TER119+ and CD71-TER119+ cell populations but not the CD71+TER119- cell population. In addition, α-toxin decreased the number of colony-forming unit erythroid colonies but not burst-forming unit erythroid colonies, indicating that α-toxin preferentially reduced mature erythroid cells compared with immature cells. α-Toxin slightly increased annexinV+ cells in TER119+ cells. Additionally, simultaneous treatment of BMCs with α-toxin and erythropoietin greatly attenuated the reduction of TER119+ erythroblasts by α-toxin. Furthermore, hemin-induced differentiation of human K562 erythroleukemia cells was impaired by α-toxin, whereas the treatment exhibited no apparent cytotoxicity. These results suggested that α-toxin mainly inhibited erythroid differentiation. Together, our results provide new insights into the biological activities of α-toxin, which might be important to understand the pathogenesis of C. perfringens infection.


Subject(s)
Bacterial Toxins/toxicity , Calcium-Binding Proteins/toxicity , Cell Differentiation/drug effects , Erythroid Precursor Cells/pathology , Erythropoiesis/drug effects , Type C Phospholipases/toxicity , Animals , Antigens, CD/metabolism , Blood Group Antigens/metabolism , Cells, Cultured , Erythroid Precursor Cells/drug effects , Humans , K562 Cells , Mice , Mice, Inbred C57BL , Receptors, Transferrin/metabolism
13.
Biochem Biophys Res Commun ; 487(2): 419-425, 2017 05 27.
Article in English | MEDLINE | ID: mdl-28419838

ABSTRACT

Granulopoiesis is accelerated during Gram-negative bacterial infection through activation of toll-like receptor 4 (TLR4). In this study, we tested whether activation of TLR2 promotes granulopoiesis by using the well-known TLR2 agonist, peptidoglycan (PGN). Neutrophils in bone marrow and spleen, and plasma granulocyte colony-stimulating factor (G-CSF) were increased in mice that had received intraperitoneal PGN administration. Incorporation of BrdU into bone marrow neutrophils increased, demonstrating that PGN accelerated granulopoiesis. Treatment of bone marrow cells (BMCs) with PGN increased neutrophils in vitro and promoted the secretion of G-CSF from Ly-6G-Ly-6C+ monocytes. The accelerated granulopoiesis caused by PGN was not seen in TLR2-deficient and MyD88-deficient BMCs. Additionally, PGN induced G-CSF production in human umbilical vein endothelial cells. These findings demonstrate that PGN promotes the secretion of G-CSF from monocytes and endothelial cells, leading to the acceleration of granulopoiesis. Our results illustrate that bacterial recognition by TLR2 facilitates granulopoiesis during Gram-positive bacterial infection.


Subject(s)
Granulocytes/physiology , Hematopoiesis/physiology , Myeloid Differentiation Factor 88/metabolism , Peptidoglycan/pharmacology , Toll-Like Receptor 2/metabolism , Animals , Cells, Cultured , Granulocytes/drug effects , Hematopoiesis/drug effects , Mice , Signal Transduction/drug effects , Signal Transduction/physiology
14.
Infect Immun ; 85(4)2017 04.
Article in English | MEDLINE | ID: mdl-28138018

ABSTRACT

Clostridium botulinum C2 toxin consists of an enzyme component (C2I) and a binding component (C2II). Activated C2II (C2IIa) binds to a cell receptor, giving rise to lipid raft-dependent oligomerization, and it then assembles with C2I. The whole toxin complex is then endocytosed into the cytosol, resulting in the destruction of the actin cytoskeleton and cell rounding. Here, we showed that C2 toxin requires acid sphingomyelinase (ASMase) activity during internalization. In this study, inhibitors of ASMase and lysosomal exocytosis blocked C2 toxin-induced cell rounding. C2IIa induced Ca2+ influx from the extracellular medium to cells. C2 toxin-induced cell rounding was enhanced in the presence of Ca2+ ASMase was released extracellularly when cells were incubated with C2IIa in the presence of Ca2+ Small interfering RNA (siRNA) knockdown of ASMase reduced C2 toxin-induced cell rounding. ASMase hydrolyzes sphingomyelin to ceramide on the outer leaflet of the membrane at acidic pH. Ceramide was detected in cytoplasmic vesicles containing C2IIa. These results indicated that ASMase activity is necessary for the efficient internalization of C2 toxin into cells. Inhibitors of ASMase may confer protection against infection.


Subject(s)
Botulinum Toxins/metabolism , Endocytosis , Sphingomyelin Phosphodiesterase/metabolism , Animals , Botulinum Toxins/toxicity , Calcium/metabolism , Cell Line , Cell Survival/drug effects , Ceramides/metabolism , Dogs , RNA Interference , RNA, Small Interfering/genetics , Sphingomyelin Phosphodiesterase/genetics
15.
Biochim Biophys Acta ; 1858(12): 3150-3156, 2016 12.
Article in English | MEDLINE | ID: mdl-27720686

ABSTRACT

BACKGROUND: Beta-toxin produced by Clostridium perfringens is a key virulence factor of fatal hemorrhagic enterocolitis and enterotoxemia. This toxin belongs to a family of ß-pore-forming toxins (PFTs). We reported recently that the ATP-gated P2X7 receptor interacts with beta-toxin. The ATP-release channel pannexin 1 (Panx1) is an important contributor to P2X7 receptor signaling. Hence, we investigated the involvement of Panx1 in beta-toxin-caused cell death. METHODS: We examined the effect of Panx1 in beta-toxin-induced cell death utilizing selective antagonists, knockdown of Panx1, and binding using dot-blot analysis. Localization of Panx1 and the P2X7 receptor after toxin treatment was determined by immunofluorescence staining. RESULTS: Selective Panx1 antagonists (carbenoxolone [CBX], probenecid, and Panx1 inhibitory peptide) prevented beta-toxin-caused cell death in THP-1 cells. CBX did not block the binding of the toxin to cells. Small interfering knockdown of Panx1 blocked beta-toxin-mediated cell death through inhibiting the oligomer formation of the toxin. Beta-toxin triggered a transient ATP release from THP-1 cells, but this early ATP release was blocked by CBX. ATP scavengers (apyrase and hexokinase) inhibited beta-toxin-induced cytotoxicity. Furthermore, co-administration of ATP with beta-toxin enhanced the binding and cytotoxicity of the toxin. CONCLUSIONS: Based on our results, Panx1 activation is achieved through the interaction of beta-toxin with the P2X7 receptor. Then, ATP released by the Panx1 channel opening promotes oligomer formation of the toxin, leading to cell death. GENERAL SIGNIFICANCE: Pannexin 1 is a novel candidate therapeutic target for beta-toxin-mediated disease.


Subject(s)
Bacterial Toxins/toxicity , Connexins/physiology , Nerve Tissue Proteins/physiology , Adenosine Triphosphate/metabolism , Apyrase/pharmacology , Carbenoxolone/pharmacology , Cell Death/drug effects , Cells, Cultured , Hexokinase/pharmacology , Humans , Receptors, Purinergic P2X7/physiology
16.
Biol Pharm Bull ; 39(10): 1694-1700, 2016.
Article in English | MEDLINE | ID: mdl-27725448

ABSTRACT

Clostridium perfringens type A, a Gram-positive, anaerobic bacterium, causes gas gangrene. Recently, we reported that C. perfringens α-toxin blocked neutrophil differentiation in an enzyme activity-dependent manner to impair host innate immunity, which should be crucial for the pathogenesis of C. perfringens. However, the detailed mechanism remains unclear. Lipid rafts have been reported to be platforms for signaling molecules involved in the regulation of cell differentiation in many different cell types. In this study, we found that cell surface expression of a lipid raft marker, GM1 ganglioside, decreased in association with neutrophil differentiation by flow cytometry analysis and morphological observation. In vitro treatment of isolated mouse bone marrow cells with α-toxin or an α-toxin variant lacking phospholipase C and sphingomyelinase activities revealed that α-toxin increased the cell surface expression of GM1 ganglioside in an enzyme activity-dependent manner. C. perfringens infection also increased GM1 ganglioside levels in bone marrow myeloid cells. Moreover, treatment of bone marrow cells with methyl-ß-cyclodextrin, a lipid raft-disrupting agent, impaired neutrophil differentiation. Together, our results suggest that the integrity of lipid rafts should be properly maintained during granulopoiesis, and α-toxin might perturb lipid raft integrity leading to the impairment of neutrophil differentiation.


Subject(s)
Bacterial Toxins/pharmacology , Bone Marrow Cells/drug effects , Calcium-Binding Proteins/pharmacology , Membrane Microdomains/drug effects , Neutrophils/drug effects , Type C Phospholipases/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , G(M1) Ganglioside/metabolism , Mice, Inbred C57BL , Neutrophils/cytology , Neutrophils/metabolism , beta-Cyclodextrins/pharmacology
17.
Sci Rep ; 6: 28192, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27306065

ABSTRACT

Although granulopoiesis is accelerated to suppress bacteria during infection, some bacteria can still cause life-threatening infections, but the mechanism behind this remains unclear. In this study, we found that mature neutrophils in bone marrow cells (BMCs) were decreased in C. perfringens-infected mice and also after injection of virulence factor α-toxin. C. perfringens infection interfered with the replenishment of mature neutrophils in the peripheral circulation and the accumulation of neutrophils at C. perfringens-infected sites in an α-toxin-dependent manner. Measurements of bacterial colony-forming units in C. perfringens-infected muscle revealed that α-toxin inhibited a reduction in the load of C. perfringens. In vitro treatment of isolated BMCs with α-toxin (phospholipase C) revealed that α-toxin directly decreased mature neutrophils. α-Toxin did not influence the viability of isolated mature neutrophils, while simultaneous treatment of BMCs with granulocyte colony-stimulating factor attenuated the reduction of mature neutrophils by α-toxin. Together, our results illustrate that impairment of the innate immune system by the inhibition of neutrophil differentiation is crucial for the pathogenesis of C. perfringens to promote disease to a life-threatening infection, which provides new insight to understand how pathogenic bacteria evade the host immune system.


Subject(s)
Bacterial Toxins/toxicity , Bone Marrow Cells/drug effects , Calcium-Binding Proteins/toxicity , Clostridium perfringens/pathogenicity , Immunity, Innate/immunology , Neutrophils/immunology , Type C Phospholipases/toxicity , Virulence Factors/toxicity , Animals , Bacillus subtilis/genetics , Bacillus subtilis/pathogenicity , Bacterial Toxins/genetics , Calcium-Binding Proteins/genetics , Cell Differentiation/drug effects , Cells, Cultured , Clostridium Infections/pathology , Clostridium perfringens/genetics , Granulocyte Colony-Stimulating Factor/pharmacology , Immunity, Innate/drug effects , Leukocyte Count , Mice , Mice, Inbred C57BL , Muscle, Skeletal/cytology , Type C Phospholipases/genetics , Virulence Factors/genetics
18.
PLoS One ; 11(1): e0147957, 2016.
Article in English | MEDLINE | ID: mdl-26807591

ABSTRACT

Clostridium perfringens delta-toxin is a ß-pore-forming toxin and a putative pathogenic agent of C. perfringens types B and C. However, the mechanism of cytotoxicity of delta-toxin remains unclear. Here, we investigated the mechanisms of cell death induced by delta-toxin in five cell lines (A549, A431, MDCK, Vero, and Caco-2). All cell lines were susceptible to delta-toxin. The toxin caused rapid ATP depletion and swelling of the cells. Delta-toxin bound and formed oligomers predominantly in plasma membrane lipid rafts. Destruction of the lipid rafts with methyl ß-cyclodextrin inhibited delta-toxin-induced cytotoxicity and ATP depletion. Delta-toxin caused the release of carboxyfluorescein from sphingomyelin-cholesterol liposomes and formed oligomers; toxin binding to the liposomes declined with decreasing cholesterol content in the liposomes. Flow cytometric assays with annexin V and propidium iodide revealed that delta-toxin treatment induced an elevation in the population of annexin V-negative and propidium iodide-positive cells. Delta-toxin did not cause the fragmentation of DNA or caspase-3 activation. Furthermore, delta-toxin caused damage to mitochondrial membrane permeability and cytochrome c release. In the present study, we demonstrate that delta-toxin produces cytotoxic activity through necrosis.


Subject(s)
Bacterial Toxins , Cell Death/drug effects , Necrosis/chemically induced , Animals , Caco-2 Cells , Caspase 3/metabolism , Chlorocebus aethiops , DNA Fragmentation/drug effects , Fluoresceins/metabolism , Humans , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Necrosis/metabolism , Vero Cells
19.
Biochim Biophys Acta ; 1850(11): 2159-67, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26299247

ABSTRACT

BACKGROUND: Clostridium perfringens beta-toxin is a pore-forming toxin (PFT) and an important agent of necrotic enteritis and enterotoxemia. We recently reported that beta-toxin strongly induced cell death in THP-1 cells via the formation of oligomers. We here describe that the P2X(7) receptor, which is an ATP receptor, interacts with beta-toxin. METHODS: We tested the role of P2X(7) receptor in beta-toxin-induced toxicity using specific inhibitors, knockdown of receptor, expression of the receptor and interaction by dot-blot assay. The potency of P2X(7) receptor was further determined using an in vivo mouse model. RESULTS: Selective P2X(7) receptor antagonists (oxidized ATP (o-ATP), oxidized ADP, and Brilliant Blue G (BBG)) inhibited beta-toxin-induced cytotoxicity in THP-1 cells. o-ATP also blocked the binding of beta-toxin to cells. The P2X(7) receptor and beta-toxin oligomer were localized in the lipid rafts of THP-1 cells. siRNA for the P2X(7) receptor inhibited toxin-induced cytotoxicity and binding of the toxin. In contrast, the siRNA knockdown of P2Y(2) or P2Y(6) had no effect on beta-toxin-induced cytotoxicity. The addition of beta-toxin to P2X(7)-transfected HEK-293 cells resulted in binding of beta-toxin oligomer. Moreover, beta-toxin specifically bound to immobilized P2X(7) receptors in vitro and colocalized with the P2X(7) receptor on the THP-1 cell surface. Furthermore, beta-toxin-induced lethality in mice was blocked by the preadministration of BBG. CONCLUSIONS: The results of this study indicate that the P2X(7) receptor plays a role in beta-toxin-mediated cellular injury. GENERAL SIGNIFICANCE: P2X(7) receptor is a potential target for the treatment of C. perfringens type C infection.


Subject(s)
Bacterial Toxins/toxicity , Receptors, Purinergic P2X7/physiology , ADAM Proteins/physiology , ADAM10 Protein , Amyloid Precursor Protein Secretases/physiology , Animals , Calcium/metabolism , HEK293 Cells , Humans , Membrane Proteins/physiology , Mice , Mice, Inbred ICR , RNA, Small Interfering/pharmacology , Rosaniline Dyes/pharmacology
20.
BMC Microbiol ; 15: 118, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26055257

ABSTRACT

BACKGROUND: Most recent studies of Clostridium perfringens plasmids have focused on toxin-encoding or antibiotic resistance plasmids. To cause intestinal disease, a toxigenic strain must grow in the intestines to levels allowing for sufficient toxin production and this in vivo growth often involves overcoming the normal intestinal microbial population. For this purpose, bacteriocin production might be important. RESULTS: In this study, as the first step in the genetic analysis of a co-existing plasmid with an enterotoxin gene (cpe)-encoding plasmid, the bacteriocin gene-encoding plasmid, pBCNF5603, was completely sequenced. This plasmid has some homology with two previously sequenced C. perfringens plasmids, namely, pCP13 carrying a cpb2 gene and pIP404 carrying a bcn gene. Using recombinant plasmids, the rep gene homologous to the PCP63 gene on pCP13 appeared to be functional. Comparative genomics indicated that the identified rep gene homologs were found on two additional toxin plasmids, pCP-OS1 and pCP-TS1. While functional analysis using recombinant plasmids indicated that pBCNF5603 and pCP13 are likely to be incompatible, the plasmid replication and partitioning region of pBCNF5603 alone was insufficient for stable maintenance of this plasmid. CONCLUSIONS: These findings suggest that pBCNF5603 evolved from recombination events between C. perfringens plasmids and inter-species mobile genetic element(s). In addition, the bcn-encoding plasmid, pBCNF5603, is likely to be included in the Inc family, which includes pCP13 and two variant iota-encoding plasmids. Furthermore, the bcn gene on pBCNF5603 could contribute to gastrointestinal disease induced by enterotoxigenic C. perfringens.


Subject(s)
Bacteriocin Plasmids/genetics , Clostridium perfringens/genetics , DNA Replication , Enterotoxins/genetics , Enterotoxins/metabolism , Molecular Sequence Data , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...