Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Anaerobe ; 82: 102753, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37308057

RESUMEN

OBJECTIVES: Clostridium perfringens epsilon-toxin is considered to be a crucial agent in enterotoxemia in domestic animals. Epsilon-toxin enters host cells via endocytosis and results in the formation of late endosome/lysosome-derived vacuoles. In the present study, we found that acid sphingomyelinase promotes the internalization of epsilon-toxin in MDCK cells. METHODS: We measured the extracellular release of acid sphingomyelinase (ASMase) by epsilon-toxin. We examined the role of ASMase in epsilon-toxin-induced cytotoxicity using selective inhibitors and knockdown of ASMase. Production of ceramide after toxin treatment was determined by immunofluorescence technique. RESULTS: Blocking agents of ASMase and exocytosis of lysosomes inhibited this epsilon-toxin-induced vacuole formation. Lysosomal ASMase was liberated to extracellular space during treatment of the cells with epsilon-toxin in the presence of Ca2+. RNAi-mediated attenuation of ASMase blocked epsilon-toxin-induced vacuolation. Moreover, incubation of MDCK cells with epsilon-toxin led to production of ceramide. The ceramide colocalized with lipid raft-binding cholera toxin subunit B (CTB) in the cell membrane, indicating that conversion of lipid raft associated sphingomyelin to ceramide by ASMase facilitates lesion of MDCK cells and internalization of epsilon-toxin. CONCLUSIONS: Based on the present results, ASMase is required for efficient internalization of epsilon-toxin.


Asunto(s)
Toxinas Bacterianas , Esfingomielina Fosfodiesterasa , Animales , Perros , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/toxicidad , Células de Riñón Canino Madin Darby , Ceramidas/metabolismo , Clostridium perfringens/metabolismo
2.
Anaerobe ; 65: 102265, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32860931

RESUMEN

Clostridium perfringens type A is the causative agent of clostridial myonecrosis, and α-toxin has been reported to be responsible for the pathogenesis. Recently, it was reported that regeneration of skeletal muscle after C. perfringens-induced muscle disorders is delayed, but the detailed mechanisms have not been elucidated. Here, we tested whether α-toxin impairs the differentiation of C2C12 myoblasts, a useful cell line to study muscle growth, maturation, and regeneration in vitro. α-Toxin dose-dependently inhibited myotube formation in C2C12 cultures after induction of their differentiation by horse serum. Also, immunoblot analysis revealed that α-toxin dose-dependently decreases the expressions of two skeletal muscle differentiation markers, myogenic differentiation 1 (MyoD) and myogenin. These results demonstrate that α-toxin impairs the myogenic differentiation of C2C12 myoblasts. To reveal the mechanism behind α-toxin-mediated impairment of myogenic differentiation, we focused on ceramide production since α-toxin is known to promote the formation of ceramide by its sphingomyelinase activity. Immunofluorescent analysis revealed that ceramide production is accelerated by treatment with α-toxin. Furthermore, a synthetic cell-permeable ceramide analog, C2-ceramide, inhibited myotube formation in C2C12 cells and decreased the expressions of MyoD and myogenin, suggesting that accelerated ceramide production is involved in the α-toxin-mediated blockage of myogenic differentiation. Together, our results illustrate that the impairment of myogenic differentiation by α-toxin might be crucial for the pathogenesis of C. perfringens to delay regeneration of severely damaged skeletal muscles.


Asunto(s)
Toxinas Bacterianas/farmacología , Proteínas de Unión al Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Mioblastos/citología , Mioblastos/efectos de los fármacos , Fosfolipasas de Tipo C/farmacología , Animales , Biomarcadores , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ratones , Desarrollo de Músculos , Proteína MioD/metabolismo , Mioblastos/metabolismo , Miogenina/metabolismo
3.
Anaerobe ; 65: 102262, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32828915

RESUMEN

Clostridium perfringens type A-induced gas gangrene is characterized by severe myonecrosis, and α-toxin has been revealed to be a major virulence factor involved in the pathogenesis. However, the detailed mechanism is unclear. Here, we show that CD31+ endothelial cell counts decrease in muscles infected with C. perfringens in an α-toxin-dependent manner. In vitro experiments revealed that α-toxin preferentially and rapidly induces the death of human umbilical vein endothelial cells (HUVECs) compared with C2C12 murine muscle cells. The toxin induces apoptosis of HUVECs by increasing ceramide. Furthermore, the specificity might be dependent on differences in the sensitivity to ceramide between these cell lines. Together, our results suggest that α-toxin-induced endothelial cell death promotes severe myonecrosis and is involved in the pathogenesis of C. perfringens.


Asunto(s)
Apoptosis , Toxinas Bacterianas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Ceramidas/metabolismo , Clostridium perfringens/fisiología , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Gangrena Gaseosa/microbiología , Fosfolipasas de Tipo C/metabolismo , Animales , Muerte Celular , Línea Celular , Células Cultivadas , Infecciones por Clostridium/metabolismo , Infecciones por Clostridium/microbiología , Clostridium perfringens/patogenicidad , Gangrena Gaseosa/metabolismo , Gangrena Gaseosa/patología , Interacciones Huésped-Patógeno , Humanos , Ratones
4.
Biochim Biophys Acta Biomembr ; 1860(2): 428-433, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28988777

RESUMEN

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.


Asunto(s)
Toxinas Bacterianas/farmacología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Proteína ADAM10/metabolismo , Células CACO-2 , Cadherinas/metabolismo , Claudina-1/metabolismo , Relación Dosis-Respuesta a Droga , Células Epiteliales/metabolismo , Humanos , Immunoblotting , Mucosa Intestinal/metabolismo , Ocludina/metabolismo , Factores de Tiempo , Proteína de la Zonula Occludens-1/metabolismo
5.
Biochim Biophys Acta ; 1858(11): 2681-2688, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27453200

RESUMEN

BACKGROUND: Clostridium perfringens epsilon-toxin is responsible for fatal enterotoxemia in ungulates. The toxin forms a heptamer in the lipid rafts of Madin-Darby Canine Kidney (MDCK) cells, leading to cell death. Here, we showed that epsilon-toxin requires neutral sphingomyelinase (nSMase) activity during oligomerization. METHODS: We tested the role of nSMase in the oligomerization of epsilon-toxin using specific inhibitors, knockdown of nSMase, formation of ceramide, and localization of epsilon-toxin and ceramide by immunofluorescence staining. RESULTS: Epsilon-toxin induced the production of ceramide is a dose- and time-dependent manner in ACHN cells. GW4869, an inhibitor of nSMase, inhibited ceramide production induced by the toxin. GW4869 and knockdown of nSMase blocked toxin-induced cell death and oligomer formation of epsilon-toxin. Confocal microscopy images showed that the toxin induced ceramide clustering and colocalized with ceramide. CONCLUSIONS: These results demonstrated that oligomer formation of epsilon-toxin is facilitated by the production of ceramide through activation of nSMase caused by the toxin. GENERAL SIGNIFICANCE: Inhibitors of nSMase may confer protection against infection.


Asunto(s)
Toxinas Bacterianas/química , Ceramidas/agonistas , Fibroblastos/enzimología , Microdominios de Membrana/efectos de los fármacos , Esfingomielina Fosfodiesterasa/metabolismo , Compuestos de Anilina/farmacología , Animales , Toxinas Bacterianas/toxicidad , Compuestos de Bencilideno/farmacología , Línea Celular , Ceramidas/biosíntesis , Clostridium perfringens/química , Perros , Activación Enzimática/efectos de los fármacos , Pruebas de Enzimas , Inhibidores Enzimáticos/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Expresión Génica , Humanos , Células de Riñón Canino Madin Darby , Microdominios de Membrana/química , Multimerización de Proteína , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/genética
6.
Biochim Biophys Acta ; 1858(12): 3150-3156, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27720686

RESUMEN

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.


Asunto(s)
Toxinas Bacterianas/toxicidad , Conexinas/fisiología , Proteínas del Tejido Nervioso/fisiología , Adenosina Trifosfato/metabolismo , Apirasa/farmacología , Carbenoxolona/farmacología , Muerte Celular/efectos de los fármacos , Células Cultivadas , Hexoquinasa/farmacología , Humanos , Receptores Purinérgicos P2X7/fisiología
7.
Infect Immun ; 85(4)2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28138018

RESUMEN

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.


Asunto(s)
Toxinas Botulínicas/metabolismo , Endocitosis , Esfingomielina Fosfodiesterasa/metabolismo , Animales , Toxinas Botulínicas/toxicidad , Calcio/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ceramidas/metabolismo , Perros , Interferencia de ARN , ARN Interferente Pequeño/genética , Esfingomielina Fosfodiesterasa/genética
8.
Biochem Biophys Res Commun ; 487(2): 419-425, 2017 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-28419838

RESUMEN

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.


Asunto(s)
Granulocitos/fisiología , Hematopoyesis/fisiología , Factor 88 de Diferenciación Mieloide/metabolismo , Peptidoglicano/farmacología , Receptor Toll-Like 2/metabolismo , Animales , Células Cultivadas , Granulocitos/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
9.
Biochim Biophys Acta ; 1850(11): 2159-67, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26299247

RESUMEN

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.


Asunto(s)
Toxinas Bacterianas/toxicidad , Receptores Purinérgicos P2X7/fisiología , Proteínas ADAM/fisiología , Proteína ADAM10 , Secretasas de la Proteína Precursora del Amiloide/fisiología , Animales , Calcio/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos ICR , ARN Interferente Pequeño/farmacología , Colorantes de Rosanilina/farmacología
10.
Biol Pharm Bull ; 39(10): 1694-1700, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27725448

RESUMEN

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.


Asunto(s)
Toxinas Bacterianas/farmacología , Células de la Médula Ósea/efectos de los fármacos , Proteínas de Unión al Calcio/farmacología , Microdominios de Membrana/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Fosfolipasas de Tipo C/farmacología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Gangliósido G(M1)/metabolismo , Ratones Endogámicos C57BL , Neutrófilos/citología , Neutrófilos/metabolismo , beta-Ciclodextrinas/farmacología
11.
BMC Microbiol ; 15: 118, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26055257

RESUMEN

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.


Asunto(s)
Plásmidos de Bacteriocinas/genética , Clostridium perfringens/genética , Replicación del ADN , Enterotoxinas/genética , Enterotoxinas/metabolismo , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Homología de Secuencia de Ácido Nucleico
12.
Biol Pharm Bull ; 38(5): 732-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25947919

RESUMEN

Clostridium perfringens TpeL belongs to a family of large clostridial glucosylating cytotoxins. TpeL modifies Rac1 and Ras subfamily proteins. Herein we report TpeL-induced formation of stress fibers via RhoA-Rho kinase (ROCK) signaling. A recombinant protein (TpeL1-525) derived from the TpeL N-terminal catalytic domain in the presence of streptolysin O (SLO) induced the formation of actin stress fibers in Madin-Darby canine kidney (MDCK) cells in a dose-dependent manner. The RhoA/ROCK pathway is known to control the formation of stress fibers. We examined the role of the RhoA/ROCK pathway in TpeL-induced formation of stress fibers. TpeL1-525-induced formation of stress fibers was inhibited by the ROCK inhibitor, Y27632 and Rho protein inhibitor, C3 transferase. TpeL1-525 activated RhoA and ROCK in a dose-dependent manner. C3 transferase blocked TpeL1-525-induced activation of RhoA and ROCK whereas Y27632 inhibited TpeL-induced activation of ROCK. These results demonstrate for the first time that TpeL induces the formation of stress fibers by activating the RhoA/ROCK signaling pathway.


Asunto(s)
Actinas/metabolismo , Toxinas Bacterianas/farmacología , Infecciones por Clostridium/metabolismo , Clostridium perfringens/patogenicidad , Fibras de Estrés/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Amidas/farmacología , Animales , Clostridium perfringens/metabolismo , Perros , Inhibidores Enzimáticos/farmacología , Células de Riñón Canino Madin Darby , Piridinas/farmacología , Transducción de Señal , Transferasas/farmacología , Proteína de Unión al GTP rac1/metabolismo , Proteínas ras/metabolismo
13.
Toxins (Basel) ; 15(12)2023 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-38133199

RESUMEN

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.


Asunto(s)
Actinas , Clostridium perfringens , Animales , Chlorocebus aethiops , Clostridium perfringens/metabolismo , Transporte Biológico , Actinas/metabolismo , Células Vero , ADP Ribosa Transferasas/química
14.
J Neurosci ; 31(14): 5225-34, 2011 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-21471357

RESUMEN

Amyloid-ß peptide (Aß) plays an important role in the pathogenesis of Alzheimer's disease (AD). Aß is generated by proteolysis of ß-amyloid precursor protein (APP) and is cleared by enzyme-mediated degradation and phagocytosis by microglia and astrocytes. Some cytokines, such as TGF-ß1, stimulate this phagocytosis. In contrast, cellular upregulation of HSP70 expression provides cytoprotection against Aß. HSP70 activity in relation to inhibition of Aß oligomerization and stimulation of Aß phagocytosis has also been reported. Although these in vitro results suggest that stimulating the expression of HSP70 could prove effective in the treatment of AD, there is a lack of in vivo evidence supporting this notion. In this study, we address this issue, using transgenic mice expressing HSP70 and/or a mutant form of APP (APPsw). Transgenic mice expressing APPsw showed less of an apparent cognitive deficit when they were crossed with transgenic mice expressing HSP70. Transgenic mice expressing HSP70 also displayed lower levels of Aß, Aß plaque deposition, and neuronal and synaptic loss than control mice. Immunoblotting experiments and direct measurement of ß- and γ-secretase activity suggested that overexpression of HSP70 does not affect the production Aß. In contrast, HSP70 overexpression did lead to upregulation of the expression of Aß-degrading enzyme and TGF-ß1 both in vivo and in vitro. These results suggest that overexpression of HSP70 in mice suppresses not only the pathological but also the functional phenotypes of AD. This study provides the first in vivo evidence confirming the potential therapeutic benefit of HSP70 for the prevention or treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas HSP70 de Choque Térmico/metabolismo , Fenotipo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Células Cultivadas , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Regulación de la Expresión Génica/genética , Proteínas HSP70 de Choque Térmico/genética , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo
15.
Biochim Biophys Acta ; 1808(4): 1103-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21255555

RESUMEN

Bacillus subtilis is a representative Gram-positive bacterium. In aerobic conditions, this bacterium can generate an electrochemical potential across the membrane with aerobic respiration. Here, we developed the patch clamp method to analyze the respiratory chain in B. subtilis. First, we prepared giant protoplasts (GPs) from B. subtilis cells. Electron micrographs and fluorescent micrographs revealed that GPs of B. subtilis had a vacuole-like structure and that the intravacuolar area was completely separated from the cytoplasmic area. Acidification of the interior of the isolated and purified vacuole-like structure, due to H(+) translocation after the addition of NADH, revealed that they consisted of everted cytoplasmic membranes. We called these giant provacuoles (GVs) and again applied the patch clamp technique. When NADH was added as an electron donor for the respiratory system, a significant NADH-induced current was observed. Inhibition of KCN and 2-heptyl-4-hydroxyquinoline-N-oxide (HQNO) demonstrated that this current is certainly due to aerobic respiration in B. subtilis. This is the first step for more detailed analyses of respiratory chain in B. subtilis, especially H(+) translocation mechanism.


Asunto(s)
Bacillus subtilis/citología , Bacillus subtilis/fisiología , Técnicas de Placa-Clamp/métodos , Bacillus subtilis/ultraestructura , Transporte de Electrón/efectos de los fármacos , Transporte de Electrón/fisiología , Hidroxiquinolinas/farmacología , Potenciales de la Membrana/efectos de los fármacos , Microscopía Electrónica de Transmisión , NAD/farmacología , Cianuro de Potasio/farmacología , Factores de Tiempo
16.
J Neurochem ; 120(5): 795-805, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22044482

RESUMEN

Amyloid-ß peptide (Aß), which is generated by the ß- and γ-secretase-mediated proteolysis of ß-amyloid precursor protein (APP), plays an important role in the pathogenesis of Alzheimer's disease (AD). We recently reported that prostaglandin E(2) (PGE(2) ) stimulates the production of Aß through both EP(2) and EP(4) receptors and that activation of the EP(4) receptor stimulates Aß production through endocytosis and activation of γ-secretase. We here found that transgenic mice expressing mutant APP (APP23) mice showed a greater or lesser apparent cognitive deficit when they were crossed with mice lacking EP(2) or EP(4) receptors, respectively. Mice lacking the EP(4) receptor also displayed lower levels of Aß plaque deposition and less neuronal and synaptic loss than control mice. Oral administration of a specific EP(4) receptor antagonist, AE3-208 to APP23 mice, improved their cognitive performance, as well as decreasing brain levels of Aß and suppressing endocytosis and activation of γ-secretase. Taken together, these results suggest that inhibition of the EP(4) receptor improves the cognitive function of APP23 mice by suppressing Aß production and reducing neuronal and synaptic loss. We therefore propose that EP(4) receptor antagonists, such as AE3-208, could be therapeutically beneficial for the prevention and treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Trastornos del Conocimiento , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Naftalenos/farmacología , Fenilbutiratos/farmacología , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/genética , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Fragmentos de Péptidos/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP4 de Receptores de Prostaglandina E/deficiencia , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
17.
Biochim Biophys Acta Biomembr ; 1864(12): 184054, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36155052

RESUMEN

Gas gangrene caused by Clostridium perfringens type A infection is a highly lethal infection of soft tissue characterized by rapid spread of tissue necrosis. This tissue destruction is related to profound attenuation of blood flow accompanied by formation of platelet-leukocyte aggregates in the blood vessels. Several studies have identified α-toxin, which has both sphingomyelinase and phospholipase C activities, as a major virulence factor in the aggregate formation via activation of the platelet gpIIbIIIa. Here, we show that α-toxin greatly and rapidly increases plasma membrane localization of CD11b, which binds to the platelet gpIIbIIIa via fibrinogen, in mouse neutrophils. Interestingly, short-term treatment of α-toxin has little effect on gene expression profiles in neutrophils, and the toxin does not change the total protein expression levels of CD11b in whole cell lysates. The following analysis demonstrated that CD11b localizes to intracellular vesicles in intact cells, but the localization changed to the cytoplasmic membrane in α-toxin-treated cells. These results suggest that CD11b is recruited to the cytoplasmic membrane by α-toxin. Previously, we reported that α-toxin promotes the formation of ceramide by its sphingomyelinase activity in mouse neutrophils. Interestingly, a synthetic cell-permeable ceramide analog, C2-ceramide, increases plasma membrane localization of CD11b, suggesting that ceramide production by α-toxin recruits CD11b to the cytoplasmic membrane to promote platelet-leukocyte aggregation. Together, our results illustrate that the increase of cell membrane CD11b expression by α-toxin might be crucial for the pathogenesis of C. perfringens to promote formation of platelet-leukocyte aggregates, leading to rapid tissue necrosis due to ischemia.


Asunto(s)
Neutrófilos , Esfingomielina Fosfodiesterasa , Animales , Toxinas Bacterianas , Antígeno CD11b , Proteínas de Unión al Calcio , Membrana Celular/metabolismo , Ceramidas , Clostridium perfringens , Fibrinógeno , Ratones , Necrosis , Neutrófilos/metabolismo , Fosfolipasas de Tipo C/metabolismo , Factores de Virulencia
18.
Case Rep Infect Dis ; 2022: 1071582, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36213874

RESUMEN

This report presents a case of a 60-year-old man who was diagnosed with ascending colon cancer with metastases of the lymph nodes and multiple liver metastases. Three days before the introduction of the first chemotherapy, he visited our hospital due to high fever. The blood test revealed an increase in the inflammatory response, hepatobiliary enzyme level, lactate dehydrogenase (LDH) level, and renal function deterioration. Contrast-enhanced computed tomography (CT) showed a rapid progression of primary lesion and liver metastatic lesions. Treatment with 5-fluorouracil, leucovorin, and oxaliplatin and cetuximab (FOLFOX/Cmab) was initiated, and the patient was admitted to our hospital after the first day of chemotherapy. At midnight, he had chills, red urine, and rapid hypoxemia. The second blood test showed progression of anemia; increased total bilirubin, aspartate aminotransferase, and LDH levels; and decreased platelet and fibrinogen levels. The serum was red wine in color, indicating marked hemolysis. The respiratory condition rapidly deteriorated, and tracheal intubation was performed and transferred into the intensive care unit. However, blood oxygenation did not increase, and the patient died the next morning, 19 h after admission, despite intensive care. Postmortem CT showed intraperitoneal free air and gas retention in the liver tumor and portal vein system. Pathological autopsy revealed perforation in ascending colon cancer, many Gram-positive rods in the perforation site, dissemination of bacteria throughout the body, and diffuse pulmonary edema. Subsequently, blood cultures reported Clostridium perfringens (CP), which is a product of alpha-toxin. CP infection can cause rapid aggravation and sudden death. The physicians should be aware of this highly fatal infection, leading to immediate diagnosis and treatment.

19.
J Biol Chem ; 285(17): 13254-63, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20177067

RESUMEN

Skin hyperpigmentation disorders due to abnormal melanin production induced by ultraviolet (UV) irradiation are both a clinical and cosmetic problem. UV irradiation stimulates melanin production in melanocytes by increasing intracellular cAMP. Expression of heat shock proteins (HSPs), especially HSP70, is induced by various stressors, including UV irradiation, to provide cellular resistance to such stressors. In this study we examined the effect of expression of HSP70 on melanin production both in vitro and in vivo. 3-Isobutyl-1-methylxanthine (IBMX), a cAMP-elevating agent, stimulated melanin production in cultured mouse melanoma cells, and this stimulation was suppressed in cells overexpressing HSP70. IBMX-dependent transcriptional activation of the tyrosinase gene was also suppressed in HSP70-overexpressing cells. Expression of microphthalmia-associated transcription factor (MITF), which positively regulates transcription of the tyrosinase gene, was up-regulated by IBMX; however, this up-regulation was not suppressed in HSP70-overexpressing cells. On the other hand, immunoprecipitation and immunostaining analyses revealed a physical interaction between and co-localization of MITF and HSP70, respectively. Furthermore, the transcription of tyrosinase gene in nuclear extract was inhibited by HSP70. In vivo, UV irradiation of wild-type mice increased the amount of melanin in the basal layer of the epidermis, and this increase was suppressed in transgenic mice expressing HSP70. This study provides the first evidence of an inhibitory effect of HSP70 on melanin production both in vitro and in vivo. This effect seems to be mediated by modulation of MITF activity through a direct interaction between HSP70 and MITF.


Asunto(s)
Regulación de la Expresión Génica , Proteínas HSP70 de Choque Térmico/metabolismo , Melaninas/biosíntesis , 1-Metil-3-Isobutilxantina/farmacología , Animales , Línea Celular Tumoral , AMP Cíclico/genética , AMP Cíclico/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Hiperpigmentación/genética , Hiperpigmentación/metabolismo , Melanoma/genética , Melanoma/metabolismo , Ratones , Ratones Transgénicos , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Monofenol Monooxigenasa/biosíntesis , Monofenol Monooxigenasa/genética , Inhibidores de Fosfodiesterasa/farmacología , Rayos Ultravioleta/efectos adversos
20.
Nihon Saikingaku Zasshi ; 76(3): 149-160, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-34789601

RESUMEN

Clostridium perfringens type A causes gas gangrene, which is a serious disease caused by wound infection. α-Toxin produced by C. perfringens is known to be the primary pathogenic factor of gas gangrene. Although it has been proposed to induce tissue damage by impairing the host immune system and peripheral circulation, sufficient findings have not been obtained to explain the high virulence of C. perfringens. For the purpose of elucidating the pathogenic mechanism of this bacterium, I focused on the disease progressions such as the bacterial colonization, muscle tissue destruction and repair, and sepsis. In this review, focusing on the action of α-toxin, it will be explained together with the latest research results that the toxin suppresses the activation of the host immune response, represents toxicity to vascular endothelial cells, induces peripheral circulatory disorders due to hematopoietic disorders, inhibits muscle tissue repair, and induces excessive immune response. These mechanisms suggest that α-toxin acts in multiple steps to disrupt host defense and that C. perfringens attacks the host with a highly sophisticated mechanism. It is expected that the onset mechanism of gas gangrene would be elucidated, and I hope that new therapeutic strategies are developed.


Asunto(s)
Toxinas Bacterianas , Gangrena Gaseosa , Proteínas de Unión al Calcio , Clostridium perfringens , Células Endoteliales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA