Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gut Microbes ; 14(1): 2122667, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36138514

RESUMEN

Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli (EHEC) cause gastrointestinal infection and, in severe cases, hemolytic uremic syndrome which may lead to death. There is, to-date, no therapy for this infection. Stx induces ATP release from host cells and ATP signaling mediates its cytotoxic effects. Apyrase cleaves and neutralizes ATP and its effect on Stx and EHEC infection was therefore investigated. Apyrase decreased bacterial RecA and dose-dependently decreased toxin release from E. coli O157:H7 in vitro, demonstrated by reduced phage DNA and protein levels. The effect was investigated in a mouse model of E. coli O157:H7 infection. BALB/c mice infected with Stx2-producing E. coli O157:H7 were treated with apyrase intraperitoneally, on days 0 and 2 post-infection, and monitored for 11 days. Apyrase-treated mice developed disease two days later than untreated mice. Untreated infected mice lost significantly more weight than those treated with apyrase. Apyrase-treated mice exhibited less colonic goblet cell depletion and apoptotic cells, as well as lower fecal ATP and Stx2, compared to untreated mice. Apyrase also decreased platelet aggregation induced by co-incubation of human platelet-rich-plasma with Stx2 and E. coli O157 lipopolysaccharide in the presence of collagen. Thus, apyrase had multiple protective effects, reducing RecA levels, stx2 and toxin release from EHEC, reducing fecal Stx2 and protecting mouse intestinal cells, as well as decreasing platelet activation, and could thereby delay the development of disease.


Asunto(s)
Bacteriófagos , Infecciones por Escherichia coli , Escherichia coli O157 , Microbioma Gastrointestinal , Adenosina Trifosfato/metabolismo , Animales , Apirasa/metabolismo , Apirasa/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/prevención & control , Escherichia coli O157/genética , Humanos , Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos BALB C , Toxina Shiga/metabolismo , Toxina Shiga/farmacología , Toxina Shiga II/genética , Toxina Shiga II/metabolismo , Toxina Shiga II/farmacología
2.
Cells ; 11(11)2022 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-35681450

RESUMEN

Shiga toxin (Stx)-producing Escherichia coli is the predominant offending agent of post-diarrheal hemolytic uremic syndrome (HUS), a rare disorder of microvascular thrombosis and acute kidney injury possibly leading to long-term renal sequelae. We previously showed that C3a has a critical role in the development of glomerular damage in experimental HUS. Based on the evidence that activation of C3a/C3a receptor (C3aR) signaling induces mitochondrial dysregulation and cell injury, here we investigated whether C3a caused podocyte and tubular injury through induction of mitochondrial dysfunction in a mouse model of HUS. Mice coinjected with Stx2/LPS exhibited glomerular podocyte and tubular C3 deposits and C3aR overexpression associated with cell damage, which were limited by C3aR antagonist treatment. C3a promoted renal injury by affecting mitochondrial wellness as demonstrated by data showing that C3aR blockade reduced mitochondrial ultrastructural abnormalities and preserved mitochondrial mass and energy production. In cultured podocytes and tubular cells, C3a caused altered mitochondrial fragmentation and distribution, and reduced anti-oxidant SOD2 activity. Stx2 potentiated the responsiveness of renal cells to the detrimental effects of C3a through increased C3aR protein expression. These results indicate that C3aR may represent a novel target in Stx-associated HUS for the preservation of renal cell integrity through the maintenance of mitochondrial function.


Asunto(s)
Síndrome Hemolítico-Urémico , Podocitos , Receptores de Complemento , Toxina Shiga II , Animales , Síndrome Hemolítico-Urémico/etiología , Síndrome Hemolítico-Urémico/metabolismo , Glomérulos Renales , Ratones , Mitocondrias/metabolismo , Podocitos/metabolismo , Receptores de Complemento/metabolismo , Toxina Shiga II/farmacología
3.
PLoS Pathog ; 17(4): e1009073, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33914852

RESUMEN

Bacteriophages (or phages) play major roles in the evolution of bacterial pathogens via horizontal gene transfer. Multiple phages are often integrated in a host chromosome as prophages, not only carrying various novel virulence-related genetic determinants into host bacteria but also providing various possibilities for prophage-prophage interactions in bacterial cells. In particular, Escherichia coli strains such as Shiga toxin (Stx)-producing E. coli (STEC) and enteropathogenic E. coli (EPEC) strains have acquired more than 10 prophages (up to 21 prophages), many of which encode type III secretion system (T3SS) effector gene clusters. In these strains, some prophages are present at a single locus in tandem, which is usually interpreted as the integration of phages that use the same attachment (att) sequence. Here, we present phages integrating into T3SS effector gene cluster-associated loci in prophages, which are widely distributed in STEC and EPEC. Some of the phages integrated into prophages are Stx-encoding phages (Stx phages) and have induced the duplication of Stx phages in a single cell. The identified attB sequences in prophage genomes are apparently derived from host chromosomes. In addition, two or three different attB sequences are present in some prophages, which results in the generation of prophage clusters in various complex configurations. These phages integrating into prophages represent a medically and biologically important type of inter-phage interaction that promotes the accumulation of T3SS effector genes in STEC and EPEC, the duplication of Stx phages in STEC, and the conversion of EPEC to STEC and that may be distributed in other types of E. coli strains as well as other prophage-rich bacterial species.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Transferencia de Gen Horizontal/genética , Profagos/genética , Toxina Shiga II/farmacología , Toxina Shiga/genética , Bacteriófagos/genética , Escherichia coli/metabolismo , Transferencia de Gen Horizontal/inmunología , Profagos/patogenicidad , Toxina Shiga II/genética , Virulencia/inmunología , Factores de Virulencia/genética
4.
Biochem Biophys Res Commun ; 557: 247-253, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33894410

RESUMEN

Accumulation of amyloid-ß peptide (Aß) in neuronal cells and in the extracellular regions in the brain is a major cause of Alzheimer's disease (AD); therefore, inhibition of Aß accumulation offers a promising approach for therapeutic strategies against AD. Aß is produced by sequential proteolysis of amyloid precursor protein (APP) in late/recycling endosomes after endocytosis of APP located in the plasma membrane. Aß is then released from cells in a free form or in an exosome-bound form. Shiga toxin (Stx) is a major virulence factor of enterohemorrhagic Escherichia coli. Recently, we found that one of the Stx subtypes, Stx2a, has a unique intracellular transport route after endocytosis through its receptor-binding B-subunit. A part of Stx2a can be transported to late/recycling endosomes and then degraded in a lysosomal acidic compartment, although in general Stx is transported to the Golgi and then to the endoplasmic reticulum in a retrograde manner. In this study, we found that treatment of APP-expressing cells with a mutant Stx2a (mStx2a), lacking cytotoxic activity because of mutations in the catalytic A-subunit, stimulated the transport of APP to the acidic compartment, which led to degradation of APP and a reduction in the amount of Aß. mStx2a-treatment also inhibited the extracellular release of Aß. Therefore, mStx2a may provide a new strategy to inhibit the production of Aß by modulating the intracellular transport of APP.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Membrana Celular/efectos de los fármacos , Endosomas/metabolismo , Lisosomas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Toxina Shiga II/farmacología , Animales , Células CHO , Dominio Catalítico/genética , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Cricetulus , Globósidos/química , Humanos , Mutación , Fosfatidilcolinas/química , Proteínas Recombinantes , Toxina Shiga II/química , Toxina Shiga II/genética , Trihexosilceramidas/química
5.
Cell Microbiol ; 22(11): e13249, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32772454

RESUMEN

Shiga toxins (Stxs) produced by Stx-producing Escherichia coli are the primarily virulence factors of hemolytic uremic syndrome and central nervous system (CNS) impairment. Although the precise mechanisms of toxin dissemination remain unclear, Stxs bind to extracellular vesicles (EVs). Exosomes, a subset of EVs, may play a key role in Stx-mediated renal injury. To test this hypothesis, we isolated exosomes from monocyte-derived macrophages in the presence of Stx2a or Stx2 toxoids. Macrophage-like differentiated THP-1 cells treated with Stxs secreted Stx-associated exosomes (Stx-Exo) of 90-130 nm in diameter, which induced cytotoxicity in recipient cells in a toxin receptor globotriaosylceramide (Gb3 )-dependent manner. Stx2-Exo engulfed by Gb3 -positive cells were translocated to the endoplasmic reticulum in the human proximal tubule epithelial cell line HK-2. Stx2-Exo contained pro-inflammatory cytokine mRNAs and proteins and induced more severe inflammation than purified Stx2a accompanied by greater death of target cells such as human renal or retinal pigment epithelial cells. Blockade of exosome biogenesis using the pharmacological inhibitor GW4869 reduced Stx2-Exo-mediated human renal cell death. Stx2-Exo isolated from human primary monocyte-derived macrophages activated caspase 3/7 and resulted in significant cell death in primary human renal cortical epithelial cells. Based on these results, we speculate that Stx-containing exosomes derived from macrophages may exacerbate cytotoxicity and inflammation and trigger cell death in toxin-sensitive cells. Therapeutic interventions targeting Stx-containing exosomes may prevent or ameliorate Stx-mediated acute vascular dysfunction.


Asunto(s)
Exosomas/metabolismo , Macrófagos/metabolismo , Toxina Shiga II/metabolismo , Toxina Shiga II/toxicidad , Trihexosilceramidas/metabolismo , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Muerte Celular , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Exosomas/inmunología , Exosomas/ultraestructura , Humanos , Inflamación , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Leucocitos Mononucleares/inmunología , Macrófagos/inmunología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Toxina Shiga II/farmacología , Células THP-1
6.
Sci Rep ; 10(1): 4945, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-32188865

RESUMEN

The cardinal virulence factor of human-pathogenic enterohaemorrhagic Escherichia coli (EHEC) is Shiga toxin (Stx), which causes severe extraintestinal complications including kidney failure by damaging renal endothelial cells. In EHEC pathogenesis, the disturbance of the kidney epithelium by Stx becomes increasingly recognised, but how this exactly occurs is unknown. To explore this molecularly, we investigated the Stx receptor content and transcriptomic profile of two human renal epithelial cell lines: highly Stx-sensitive ACHN cells and largely Stx-insensitive Caki-2 cells. Though both lines exhibited the Stx receptor globotriaosylceramide, RNAseq revealed strikingly different transcriptomic responses to an Stx challenge. Using RNAi to silence factors involved in ACHN cells' Stx response, the greatest protection occurred when silencing RAB5A and TRAPPC6B, two host factors that we newly link to Stx trafficking. Silencing these factors alongside YKT6 fully prevented the cytotoxic Stx effect. Overall, our approach reveals novel subcellular targets for potential therapies against Stx-mediated kidney failure.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Riñón/efectos de los fármacos , Toxina Shiga II/farmacología , Proteínas de Transporte Vesicular/antagonistas & inhibidores , Proteínas de Unión al GTP rab5/antagonistas & inhibidores , Células Cultivadas , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Humanos , Riñón/metabolismo
7.
PLoS Pathog ; 15(10): e1008003, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31581229

RESUMEN

Specific Escherichia coli isolates lysogenised with prophages that express Shiga toxin (Stx) can be a threat to human health, with cattle being an important natural reservoir. In many countries the most severe pathology is associated with enterohaemorrhagic E. coli (EHEC) serogroups that express Stx subtype 2a. In the United Kingdom, phage type (PT) 21/28 O157 strains have emerged as the predominant cause of life-threatening EHEC infections and this phage type commonly encodes both Stx2a and Stx2c toxin types. PT21/28 is also epidemiologically linked to super-shedding (>103 cfu/g of faeces) which is significant for inter-animal transmission and human infection as demonstrated using modelling studies. We demonstrate that Stx2a is the main toxin produced by stx2a+/stx2c+ PT21/28 strains induced with mitomycin C and this is associated with more rapid induction of gene expression from the Stx2a-encoding prophage compared to that from the Stx2c-encoding prophage. Bacterial supernatants containing either Stx2a and/or Stx2c were demonstrated to restrict growth of bovine gastrointestinal organoids with no restriction when toxin production was not induced or prevented by mutation. Isogenic strains that differed in their capacity to produce Stx2a were selected for experimental oral colonisation of calves to assess the significance of Stx2a for both super-shedding and transmission between animals. Restoration of Stx2a expression in a PT21/28 background significantly increased animal-to-animal transmission and the number of sentinel animals that became super-shedders. We propose that while both Stx2a and Stx2c can restrict regeneration of the epithelium, it is the relatively rapid and higher levels of Stx2a induction, compared to Stx2c, that have contributed to the successful emergence of Stx2a+ E. coli isolates in cattle in the last 40 years. We propose a model in which Stx2a enhances E. coli O157 colonisation of in-contact animals by restricting regeneration and turnover of the colonised gastrointestinal epithelium.


Asunto(s)
Enfermedades de los Bovinos/transmisión , Células Epiteliales/microbiología , Infecciones por Escherichia coli/veterinaria , Escherichia coli O157/efectos de los fármacos , Íleon/microbiología , Organoides/microbiología , Toxina Shiga II/farmacología , Animales , Bovinos , Enfermedades de los Bovinos/epidemiología , Enfermedades de los Bovinos/microbiología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Escherichia coli O157/aislamiento & purificación , Íleon/citología , Íleon/metabolismo , Masculino , Organoides/crecimiento & desarrollo , Organoides/metabolismo , Virulencia
8.
Metabolomics ; 15(10): 131, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31576432

RESUMEN

INTRODUCTION: Shiga toxin 2a (Stx2a) induces hemolytic uremic syndrome (STEC HUS) by targeting glomerular endothelial cells (GEC). OBJECTIVES: We investigated in a metabolomic analysis the response of a conditionally immortalized, stable glomerular endothelial cell line (ciGEnC) to Stx2a stimulation as a cell culture model for STEC HUS. METHODS: CiGEnC were treated with tumor necrosis factor-(TNF)α, Stx2a or sequentially with TNFα and Stx2a. We performed a metabolomic high-throughput screening by lipid- or gas chromatography and subsequent mass spectrometry. Metabolite fold changes in stimulated ciGEnC compared to untreated cells were calculated. RESULTS: 320 metabolites were identified and investigated. In response to TNFα + Stx2a, there was a predominant increase in intracellular free fatty acids and amino acids. Furthermore, lipid- and protein derived pro-inflammatory mediators, oxidative stress and an augmented intracellular energy turnover were increased in ciGEnC. Levels of most biochemicals related to carbohydrate metabolism remained unchanged. CONCLUSION: Stimulation of ciGEnC with TNFα + Stx2a is associated with profound metabolic changes indicative of increased inflammation, oxidative stress and energy turnover.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Glomérulos Renales/citología , Metabolómica , Toxina Shiga II/farmacología , Recuento de Células , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/citología , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos , Análisis Multivariante , Toxina Shiga II/metabolismo
9.
Biol Pharm Bull ; 41(9): 1475-1479, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30175782

RESUMEN

Shiga toxin (Stx) is a main virulence factor of Enterohemorrhagic Escherichia coli (EHEC) that causes diarrhea and hemorrhagic colitis and occasionally fatal systemic complications. Stx induces rapid apoptotic cell death in some cells, such as human myelogenous leukemia THP-1 cells expressing CD77, a receptor for Stx internalization, and the induction of apoptotic cell death is thought to be crucial for the fatal systemic complications. Therefore, in order to suppress the fatal toxicity, it is important to understand the mechanism how cells can escape from apoptotic cell death in the presence of Stx. In this study, we isolated resistant clones to Stx-induced apoptosis from highly sensitive THP-1 cells by continuous exposure with lethal dose of Stx. All of the ten resistant clones lost the expression of CD77 as a consequence of the reduction in CD77 synthase mRNA expression. These results suggest that downregulation of CD77 or CD77 synthase expression could be a novel approach to suppress the fatal toxicity of Stx in EHEC infected patient.


Asunto(s)
Galactosiltransferasas/genética , Leucemia Mieloide/metabolismo , Toxina Shiga I/farmacología , Toxina Shiga II/farmacología , Trihexosilceramidas/metabolismo , Antineoplásicos Fitogénicos/farmacología , Apoptosis , Etopósido/farmacología , Humanos , Células THP-1
10.
Am J Physiol Renal Physiol ; 315(4): F861-F869, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29513070

RESUMEN

Endothelial injury with consecutive microangiopathy and endothelial dysfunction plays a central role in the pathogenesis of the postenteropathic hemolytic uremic syndrome (D + HUS). To identify new treatment strategies, we examined the regenerative potential of endothelial progenitor cells (EPCs) in an in vitro model of Shiga toxin (Stx) 2a-induced glomerular endothelial injury present in D + HUS and the mechanisms of EPC-triggered endothelial regeneration. We simulated the proinflammatory milieu present in D + HUS by priming human renal glomerular endothelial cells (HRGECs) with tumor necrosis factor-α before stimulation with Stx2a. This measure led to a time- and concentration-dependent decrease of HRGEC viability of human renal glomerular endothelial cells as detected by a colorimetric assay. Coincubation with EPCs (104-105 cells/ml) under dynamic flow conditions led to a significant improvement of cell viability in comparison to untreated monolayers (0.45 ± 0.06 vs. 0.16 ± 0.04, P = 0.003). A comparable regenerative effect of EPCs was observed in a coculture model using cell culture inserts (0.41 ± 0.05 vs. 0.16 ± 0.04, P = 0.003) associated with increased concentrations of vascular endothelial growth factor, insulin-like growth factor I, fibroblast growth factor-2, and hepatocyte growth factor in the supernatant. Treatment of Stx2a-injured monolayers with a combination of these growth factors imitated this effect. EPCs did not show distinct sings of migration and angiogenic tube formation in functional assays. These data demonstrate that EPCs significantly improve endothelial viability after Stx2a-induced injury in vitro and that this effect is associated with the release of growth factors by EPCs.


Asunto(s)
Células Progenitoras Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Regeneración/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Progenitoras Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Humanos , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Toxina Shiga II/farmacología , Células Madre/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos
11.
Sci Rep ; 5: 17479, 2015 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-26626573

RESUMEN

Infection with Escherichia coli O157:H7 may develop into hemorrhagic colitis, or hemolytic uremic syndrome (HUS), which usually causes kidney failure or even death. The adhesion and toxins are the important virulent factors. In this study, a novel vaccine candidate rSOBGs was constructed based on the bacterial ghost (BG). rSOBGs maintained the integrity of cellular morphology and displayed the linear Stx2Am-Stx1B antigen on the surface of outer membrane. rSOBGs induced Stxs-specific IgA/IgG antibodies and stronger intimin-specific IgA/IgG antibodies effectively in sera in this study. In vivo, the rSOBGs provided the higher protection rate (52%) than native bacterial ghost-OBGs (12%) when challenged intragastricly with high dose (500 LD50) viable E. coli O157:H7. Meanwhile, the rSOBGs provided higher protection rate (73.33%) than OBGs when challenged with 2 LD50 even to 5 LD50 lysed E. coli O157:H7. In vitro, the rSOBGs-immunized sera possessed neutralizing activity to lysed pathogenic bacteria. Furthermore, the results of histopathology also displayed that the administration of rSOBGs have the ability to reduce or inhibit the adhesion lesions and toxins damages of organs. The novel vaccine candidate rSOBGs induced both anti-toxin and anti-adhesion immune protection, suggesting the possibility to prevent the infectious diseases caused by Escherichia coli O157:H7.


Asunto(s)
Antígenos Bacterianos/farmacología , Escherichia coli O157/inmunología , Vacunas contra Escherichia coli/farmacología , Síndrome Hemolítico-Urémico/prevención & control , Toxina Shiga I/farmacología , Toxina Shiga II/farmacología , Animales , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Escherichia coli O157/genética , Vacunas contra Escherichia coli/genética , Vacunas contra Escherichia coli/inmunología , Síndrome Hemolítico-Urémico/genética , Síndrome Hemolítico-Urémico/inmunología , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología , Toxina Shiga I/genética , Toxina Shiga I/inmunología , Toxina Shiga II/genética , Toxina Shiga II/inmunología
12.
Cell Biol Toxicol ; 30(5): 289-99, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25200685

RESUMEN

The blood-brain barrier (BBB) selectively controls the homeostasis of the central nervous system (CNS) environment using specific structural and biochemical features of the endothelial cells, pericytes, and glial limitans. Glial cells, which represent the cellular components of the mature BBB, are the most numerous cells in the brain and are indispensable for neuronal functioning. We investigated the effects of Shiga toxin on glial cells in vitro. Shiga toxin failed to inhibit cell proliferation but attenuated expression of heat shock protein 70, which is one of the chaperone proteins, in cultured and primary glial cells. Furthermore, the combination of Shiga toxin and a heat shock procedure induced cell apoptosis and decreased cell proliferation in both cells. Thus, we speculate that glial cell death in response to the combination of Shiga toxin and heat shock might weaken the BBB and induce central nervous system complications.


Asunto(s)
Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Neuroglía/efectos de los fármacos , Toxina Shiga II/farmacología , Animales , Barrera Hematoencefálica/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Calor , Ratas
13.
J Am Soc Nephrol ; 25(8): 1786-98, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24578132

RESUMEN

Shiga toxin (Stx)-producing Escherichia coli is the offending agent of postdiarrhea-associated hemolytic uremic syndrome (HUS), a disorder of glomerular ischemic damage and widespread microvascular thrombosis. We previously documented that Stx induces glomerular complement activation, generating C3a responsible for microvascular thrombosis in experimental HUS. Here, we show that the presence of C3 deposits on podocytes is associated with podocyte damage and loss in HUS mice generated by the coinjection of Stx2 and LPS. Because podocyte adhesion to the glomerular basement membrane is mediated by integrins, the relevance of integrin-linked kinase (ILK) signals in podocyte dysfunction was evaluated. Podocyte expression of ILK increased after the injection of Stx2/LPS and preceded the upregulation of Snail and downregulation of nephrin and α-actinin-4. Factor B deficiency or pretreatment with an inhibitory antibody to factor B protected mice against Stx2/LPS-induced podocyte dysregulation. Similarly, pretreatment with a C3a receptor antagonist limited podocyte loss and changes in ILK, Snail, and α-actinin-4 expression. In cultured podocytes, treatment with C3a reduced α-actinin-4 expression and promoted ILK-dependent nuclear expression of Snail and cell motility. These results suggest that Stx-induced activation of the alternative pathway of complement and generation of C3a promotes ILK signaling, leading to podocyte dysfunction and loss in Stx-HUS.


Asunto(s)
Complemento C3a/metabolismo , Vía Alternativa del Complemento/efectos de los fármacos , Síndrome Hemolítico-Urémico/patología , Glomérulos Renales/efectos de los fármacos , Podocitos/efectos de los fármacos , Toxina Shiga II/farmacología , Animales , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Síndrome Hemolítico-Urémico/etiología , Síndrome Hemolítico-Urémico/metabolismo , Humanos , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Podocitos/metabolismo , Podocitos/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal
14.
PLoS One ; 8(7): e70431, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936204

RESUMEN

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.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Proteínas de Escherichia coli/farmacología , Glomérulos Renales/efectos de los fármacos , Toxina Shiga II/farmacología , Subtilisinas/farmacología , Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Proteínas de Escherichia coli/toxicidad , Humanos , Glomérulos Renales/metabolismo , Necrosis/tratamiento farmacológico , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Toxina Shiga II/toxicidad , Subtilisinas/toxicidad , Factor de von Willebrand/metabolismo
15.
J Am Soc Nephrol ; 24(9): 1413-23, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23744887

RESUMEN

Hemolytic uremic syndrome, a life-threatening disease often accompanied by acute renal failure, usually occurs after gastrointestinal infection with Shiga toxin 2 (Stx2)-producing Escherichia coli. Stx2 binds to the glycosphingolipid globotriaosylceramide receptor, expressed by renal epithelial cells, and triggers apoptosis by activating the apoptotic factor Bax. Signaling via the ouabain/Na,K-ATPase/IP3R/NF-κB pathway increases expression of Bcl-xL, an inhibitor of Bax, suggesting that ouabain might protect renal cells from Stx2-triggered apoptosis. Here, exposing rat proximal tubular cells to Stx2 in vitro resulted in massive apoptosis, upregulation of the apoptotic factor Bax, increased cleaved caspase-3, and downregulation of the survival factor Bcl-xL; co-incubation with ouabain prevented all of these effects. Ouabain activated the NF-κB antiapoptotic subunit p65, and the inhibition of p65 DNA binding abolished the antiapoptotic effect of ouabain in Stx2-exposed tubular cells. Furthermore, in vivo, administration of ouabain reversed the imbalance between Bax and Bcl-xL in Stx2-treated mice. Taken together, these results suggest that ouabain can protect the kidney from the apoptotic effects of Stx2.


Asunto(s)
Apoptosis/efectos de los fármacos , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/fisiopatología , Ouabaína/farmacología , Toxina Shiga II/farmacología , Proteína X Asociada a bcl-2/fisiología , Proteína bcl-X/fisiología , Animales , Apoptosis/fisiología , Caspasa 3/fisiología , Caspasa 8/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Técnicas In Vitro , Receptores de Inositol 1,4,5-Trifosfato/fisiología , Túbulos Renales Proximales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/fisiología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , ATPasa Intercambiadora de Sodio-Potasio/fisiología , Proteína X Asociada a bcl-2/efectos de los fármacos , Proteína bcl-X/efectos de los fármacos
16.
Glycobiology ; 23(6): 745-59, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23431059

RESUMEN

Shiga toxin (Stx) 2e, released by certain Stx-producing Escherichia coli, is presently the best characterized virulence factor responsible for pig edema disease, which is characterized by hemorrhagic lesions, neurological disorders and often fatal outcomes. Although Stx2e-mediated brain vascular injury is the key event in development of neurologic signs, the glycosphingolipid (GSL) receptors of Stx2e and toxin-mediated impairment of pig brain endothelial cells have not been investigated so far. Here, we report on the detailed structural characterization of Stx2e receptors globotriaosylceramide (Gb3Cer) and globotetraosylceramide (Gb4Cer), which make up the major neutral GSLs in primary porcine brain capillary endothelial cells (PBCECs). Various Gb3Cer and Gb4Cer lipoforms harboring sphingenine (d18:1) or sphinganine (d18:0) and mostly a long-chain fatty acid (C20-C24) were detected. A notable batch-to-batch heterogeneity of primary endothelial cells was observed regarding the extent of ceramide hydroxylation of Gb3Cer or Gb4Cer species. Gb3Cer, Gb4Cer and sphingomyelin preferentially distribute to detergent-resistant membrane fractions and can be considered lipid raft markers in PBCECs. Moreover, we employed an in vitro model of the blood-brain barrier (BBB), which exhibited strong cytotoxic effects of Stx2e on the endothelial monolayer and a rapid collapse of the BBB. These data strongly suggest the involvement of Stx2e in cerebral vascular damage with resultant neurological disturbance characteristic of edema disease.


Asunto(s)
Barrera Hematoencefálica/patología , Células Endoteliales/metabolismo , Globósidos/metabolismo , Trihexosilceramidas/metabolismo , Animales , Barrera Hematoencefálica/inmunología , Encéfalo/patología , Conformación de Carbohidratos , Secuencia de Carbohidratos , Técnicas de Cultivo de Célula , Membrana Celular/metabolismo , Células Cultivadas , Impedancia Eléctrica , Células Endoteliales/inmunología , Endotelio/inmunología , Endotelio/fisiopatología , Globósidos/química , Glucolípidos/química , Glucolípidos/metabolismo , Datos de Secuencia Molecular , Cultivo Primario de Células , Receptores de Superficie Celular/química , Receptores de Superficie Celular/metabolismo , Toxina Shiga II/farmacología , Sus scrofa , Trihexosilceramidas/química
17.
Toxins (Basel) ; 4(12): 1517-34, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23242319

RESUMEN

Our previous genetic, pharmacological and analogue protection studies identified the glycosphingolipid, Gb(3) (globotriaosylceramide, Pk blood group antigen) as a natural resistance factor for HIV infection. Gb(3) is a B cell marker (CD77), but a fraction of activated peripheral blood mononuclear cells (PBMCs) can also express Gb(3). Activated PBMCs predominantly comprise CD4+ T-cells, the primary HIV infection target. Gb(3) is the sole receptor for Escherichia coli verotoxins (VTs, Shiga toxins). VT1 contains a ribosome inactivating A subunit (VT1A) non-covalently associated with five smaller receptor-binding B subunits. The effect of VT on PHA/IL2-activated PBMC HIV susceptibility was determined. Following VT1 (or VT2) PBMC treatment during IL2/PHA activation, the small Gb(3)+/CD4+ T-cell subset was eliminated but, surprisingly, remaining CD4+ T-cell HIV-1(IIIB) (and HIV-1(Ba-L)) susceptibility was significantly reduced. The Gb(3)-Jurkat T-cell line was similarly protected by brief VT exposure prior to HIV-1(IIIB) infection. The efficacy of the VT1A subunit alone confirmed receptor independent protection. VT1 showed no binding or obvious Jurkat cell/PBMC effect. Protective VT1 concentrations reduced PBMC (but not Jurkat cell) proliferation by 50%. This may relate to the mechanism of action since HIV replication requires primary T-cell proliferation. Microarray analysis of VT1A-treated PBMCs indicated up regulation of 30 genes. Three of the top four were histone genes, suggesting HIV protection via reduced gene activation. VT blocked HDAC inhibitor enhancement of HIV infection, consistent with a histone-mediated mechanism. We speculate that VT1A may provide a benign approach to reduction of (X4 or R5) HIV cell susceptibility.


Asunto(s)
Infecciones por VIH/prevención & control , Subunidades de Proteína/farmacología , Toxina Shiga I/farmacología , Toxina Shiga II/farmacología , Linfocitos T/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Perfilación de la Expresión Génica , VIH-1/efectos de los fármacos , VIH-1/patogenicidad , Humanos , Células Jurkat , Leucocitos Mononucleares , Análisis de Secuencia por Matrices de Oligonucleótidos
18.
Biol Chem ; 393(8): 785-99, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22944681

RESUMEN

Shiga toxins (Stxs) are composed of an enzymatically active A subunit (StxA) and a pentameric B subunit (StxB) that preferentially binds to the glycosphingolipid (GSL) globo\xadtriaosylceramide (Gb3Cer/CD77) and to a reduced extent to globotetraosylceramide (Gb4Cer). The identification of Gb3Cer as a tumor-associated GSL in human pancreatic cancer prompted us to investigate the expression of Gb3Cer and Gb4Cer in 15 human pancreatic ductal adenocarcinoma cell lines derived from primary tumors and liver, ascites, and lymph node metastases. Thin-layer chromatography overlay assays revealed the occurrence of Gb3Cer in all and of Gb4Cer in the majority of cell lines, which largely correlated with transcriptional expression analysis of Gb3Cer and Gb4Cer synthases. Prominent Gb3Cer and Gb4Cer lipoform heterogeneity was based on ceramides carrying predominantly C16:0 and C24:0/C24:1 fatty acids. Stx2-mediated cell injury ranged from extremely high sensitivity (CD(50) of 0.94 pg/ml) to high refractiveness (CD(50) of 5.8 µg/ml) and to virtual resistance portrayed by non-determinable CD(50) values even at the highest Stx2 concentration (10 µg/ml) applied. Importantly, Stx2-mediated cytotoxicity did not correlate with Gb3Cer expression (the preferential Stx receptor), suggesting that the GSL receptor content does not primarily determine cell sensitivity and that other, yet to be delineated, cellular factors might influence the responsiveness of cancer cells.


Asunto(s)
Adenocarcinoma/genética , Carcinoma Ductal Pancreático/genética , Supervivencia Celular/efectos de los fármacos , Globósidos/genética , Toxina Shiga II/farmacología , Trihexosilceramidas/genética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Adenocarcinoma/secundario , Ascitis/patología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/secundario , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Globósidos/análisis , Globósidos/metabolismo , Humanos , Neoplasias Hepáticas/patología , Ganglios Linfáticos/patología , Toxina Shiga II/aislamiento & purificación , Escherichia coli Shiga-Toxigénica/química , Trihexosilceramidas/análisis , Trihexosilceramidas/metabolismo
19.
Sci Rep ; 2: 631, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22953052

RESUMEN

Shiga-like toxins and ricin are ribosome-inactivating proteins (RIPs) that are lethal to mammals and pose a global health threat. No clinical vaccines or therapeutics currently exist to protect against these RIPs. Two small molecules (Retro-1 and Retro-2) were discovered with high-throughput screening and reported for their protection of cells against RIPs. Of great significance, Retro-2, reported as (E)-2-(((5-methylthiophen-2-yl)methylene)amino)-N-phenylbenzamide, fully protected mice from lethal nasal challenge with ricin. Herein, we report studies showing that the chemical structure of Retro-2 is (±)-2-(5-methylthiophen-2-yl)-3-phenyl-2,3-dihydroquinazolin-4(1H)-one rather than (E)-2-(((5-methylthiophen-2-yl)methylene)amino)-N-phenylbenzamide. The latter is an achiral molecule that converts spontaneously to the former, which is a racemate and showed cell protection against RIPs. This calls for attention to (±)-2-(5-methylthiophen-2-yl)-3-phenyl-2,3-dihydroquinazolin-4(1H)-one as a promising RIP inhibitor and for chemical characterization of drug leads obtained from high-throughput screens.


Asunto(s)
Benzamidas/química , Ricina/farmacología , Toxina Shiga II/farmacología , Tiofenos/química , Animales , Benzamidas/síntesis química , Benzamidas/farmacología , Chlorocebus aethiops , Citoprotección , Estabilidad de Medicamentos , Espectroscopía de Resonancia Magnética , Metanol/química , Estructura Molecular , Proteínas Inactivadoras de Ribosomas , Solventes/química , Estereoisomerismo , Tiofenos/síntesis química , Tiofenos/farmacología , Células Vero
20.
Blood ; 120(5): 1143-9, 2012 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-22718838

RESUMEN

Shiga toxin (Stx) causes diarrhea-associated hemolytic uremic syndrome by damaging renal microvascular endothelium. The pentameric B subunits of Stx types 1 and 2 (Stx1B and Stx2B) are sufficient to stimulate acute VWF secretion from endothelial cells, but Stx1B and Stx2B exert distinct effects on Ca(2+) and cAMP pathways. Therefore, we investigated other signaling components in StxB-induced VWF exocytosis. Incubation of HUVECs with StxB transiently increased phospholipase D (PLD) activity. Inhibition of PLD activity or shRNA-mediated PLD1 knockdown abolished StxB-induced VWF secretion. In addition, treatment with StxB triggered actin polymerization, enhanced endothelial monolayer permeability, and activated RhoA. PLD activation and VWF secretion induced by Stx1B were abolished on protein kinase Cα (PKCα) inhibition or gene silencing but were only moderately reduced by Rho or Rho kinase inhibitors. Conversely, PLD activation and VWF exocytosis induced by Stx2B were reduced by Rho/Rho kinase inhibitors and dominant-negative RhoA, whereas attenuation of PKCα did not affect either process. Another PLD1 activator, ADP-ribosylation factor 6, was involved in VWF secretion induced by Stx1B or Stx2B, but not histamine. These data indicate that Stx1B and Stx2B induce acute VWF secretion in a PLD1-dependent manner but do so by differentially modulating PKCα, RhoA, and ADP-ribosylation factor 6.


Asunto(s)
Fosfolipasa D/fisiología , Toxinas Shiga/farmacología , Factor de von Willebrand/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Células Cultivadas , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Permeabilidad/efectos de los fármacos , Fosfolipasa D/antagonistas & inhibidores , Fosfolipasa D/genética , Fosfolipasa D/metabolismo , Proteína Quinasa C-alfa/metabolismo , Subunidades de Proteína/metabolismo , Subunidades de Proteína/farmacología , ARN Interferente Pequeño/farmacología , Toxina Shiga II/farmacología , Toxinas Shiga/química , Factores de Tiempo , Proteína de Unión al GTP rhoA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...