Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
Int J Mol Sci ; 22(9)2021 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-34066520

RESUMEN

Shiga toxin (STx) or Vero toxin is a virulence factor produced by enterohemorrhagic Escherichia coli. The toxin binds to the glycosphingolipid globotriaosylceramide (Gb3) for its entry, and causes cell death by inhibiting ribosome function. Previously, we performed a loss-of-function screen in HeLa cells using a human CRISPR knockout (KO) library and identified various host genes required for STx-induced cell death. To determine whether this library targeted to the human genome is applicable to non-human primate cells and to identify previously unrecognized factors crucial for STx-induced cell death, we herein performed a similar screen in the African green monkey kidney-derived Vero C1008 subline. Many genes relevant to metabolic enzymes and membrane trafficking were enriched, although the number of enriched genes was less than that obtained in the screening for HeLa cells. Of note, several genes that had not been enriched in the previous screening were enriched: one of these genes was SYS1, which encodes a multi-spanning membrane protein in the Golgi apparatus. In SYS1 KO Vero cells, expression of Gb3 and sphingomyelin was decreased, while that of glucosylceramide and lactosylceramide was increased. In addition, loss of SYS1 inhibited the biosynthesis of protein glycans, deformed the Golgi apparatus, and perturbed the localization of trans-Golgi network protein (TGN) 46. These results indicate that the human CRISPR KO library is applicable to Vero cell lines, and SYS1 has a widespread effect on glycan biosynthesis via regulation of intra-Golgi and endosome-TGN retrograde transports.


Asunto(s)
Proteínas de la Membrana/metabolismo , Toxina Shiga/toxicidad , Secuencia de Aminoácidos , Animales , Sistemas CRISPR-Cas/genética , Muerte Celular/efectos de los fármacos , Chlorocebus aethiops , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Humanos , Proteínas de la Membrana/química , Polisacáridos/biosíntesis , Proteínas de Unión al ARN/metabolismo , Células Vero , Red trans-Golgi/metabolismo
2.
Nephrol Dial Transplant ; 35(12): 2147-2153, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-31411695

RESUMEN

BACKGROUND: Treatment with eculizumab in Shiga toxin-associated haemolytic and uraemic syndrome (STEC-HUS) remains controversial despite its increasing utilization. The aim of our study was to evaluate the outcomes of children treated with eculizumab for STEC-HUS in a single-centre matched cohort study. METHODS: Data were retrospectively collected from medical records of children diagnosed with STEC-HUS. The outcomes of patients treated with eculizumab for STEC-HUS were compared with those of a control group of untreated patients matched for age, sex and severity of acute kidney injury with a 1:2 matching scheme. RESULTS: Eighteen children (median age 40.6 months) with STEC-HUS treated with eculizumab were compared with 36 matched control patients (median age 36.4 months) who did not receive eculizumab. All patients survived in the two groups. Within 1 month of HUS onset, the evolution of haematological and renal parameters did not differ between the two groups. At 12 months of follow-up, renal outcome was not significantly different between the two groups. At the last follow-up, the prevalence of decreased glomerular filtration rate in the eculizumab group (27%) was not statistically different from that in controls (38%), as was the prevalence of proteinuria and high blood pressure. Children who received eculizumab more often had extrarenal sequelae during follow-up. Eculizumab treatment appeared to be safe in children with STEC-HUS. CONCLUSION: The benefit of eculizumab on renal and extrarenal outcomes in STEC-HUS could not be established based on our findings. However, efficacy and safety are not best assessed by the observational design and small sample size of our study. Randomized controlled trials are thus required to determine the efficacy of eculizumab in this indication.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Inactivadores del Complemento/uso terapéutico , Infecciones por Escherichia coli/complicaciones , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Toxina Shiga/toxicidad , Escherichia coli Shiga-Toxigénica/patogenicidad , Estudios de Casos y Controles , Niño , Preescolar , Infecciones por Escherichia coli/inducido químicamente , Infecciones por Escherichia coli/microbiología , Femenino , Síndrome Hemolítico-Urémico/microbiología , Síndrome Hemolítico-Urémico/patología , Humanos , Lactante , Masculino , Estudios Retrospectivos , Resultado del Tratamiento
3.
Pediatr Nephrol ; 34(3): 379-388, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-29214442

RESUMEN

Shiga toxin (Stx)-producing Escherichia coli (STEC) is the offending agent in post-diarrhea-associated hemolytic uremic syndrome (HUS), a disorder characterized by thrombocytopenia, microangiopathic hemolytic anemia, and acute kidney failure, with thrombi occluding the renal microvasculature. Endothelial dysfunction has been recognized as the trigger event in the development of microangiopathic processes. Glomerular endothelial cells are susceptible to the toxic effects of Stxs that, via nuclear factor kappa B (NF-κB) activation, induce the expression of genes encoding for adhesion molecules and chemokines, culminating in leukocyte adhesion and platelet thrombus formation on the activated endothelium. Complement activation via the alternative pathway has been seen in patients during the acute phase of STEC-associated HUS. Experimental evidence has highlighted the role of complement proteins in driving glomerular endothelium toward a thrombogenic phenotype. At the glomerular level, podocytes are also an important target of Stx-induced complement activation. Glomerular injury as a consequence of podocyte dysfunction and loss is thus a mechanism that might affect long-term renal outcomes in the disease. New approaches to targeting the complement system may be useful therapeutic options for patients with STEC-HUS.


Asunto(s)
Células Endoteliales/patología , Síndrome Hemolítico-Urémico/inmunología , Podocitos/patología , Toxina Shiga/toxicidad , Escherichia coli Shiga-Toxigénica/patogenicidad , Animales , Colon/microbiología , Vía Alternativa del Complemento/efectos de los fármacos , Vía Alternativa del Complemento/inmunología , Diarrea/complicaciones , Diarrea/microbiología , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Síndrome Hemolítico-Urémico/microbiología , Síndrome Hemolítico-Urémico/patología , Humanos , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Mucosa Intestinal/microbiología , Microvasos/citología , Microvasos/inmunología , Microvasos/patología , Podocitos/inmunología , Toxina Shiga/inmunología , Toxina Shiga/metabolismo , Escherichia coli Shiga-Toxigénica/inmunología , Escherichia coli Shiga-Toxigénica/metabolismo
4.
J Pediatr Hematol Oncol ; 41(3): e179-e181, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29683956

RESUMEN

Diarrhea-associated hemolytic uremic syndrome (HUS) is usually associated with shigatoxin-producing Escherichia coli or shigella infections. We report 2 cases of HUS, respectively, caused by salmonella and Campylobacter jejuni infections. None of these bacteria produce shigatoxins, and the underlying mechanism of HUS development remains unknown. In streptococcus pneumoniae-associated HUS, bacterial neuraminidase cleaves neuraminic acid and causes exposure of Thomsen-Friedenreich cryptantigen on the cell surface of, for example, erythrocytes, which induces an inflammatory response caused by binding of preformed IgM. Both campylobacter and salmonella bacteria also produce neuraminidase, and HUS development could be explained by a similar mechanism.


Asunto(s)
Campylobacter jejuni/patogenicidad , Síndrome Hemolítico-Urémico/microbiología , Salmonella/patogenicidad , Infecciones por Campylobacter/complicaciones , Síndrome Hemolítico-Urémico/etiología , Humanos , Neuraminidasa , Infecciones por Salmonella/complicaciones , Toxina Shiga/toxicidad
5.
J Lipid Res ; 59(8): 1383-1401, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29866658

RESUMEN

Shiga toxins (Stxs) are the major virulence factors of Stx-producing Escherichia coli (STEC), which cause hemorrhagic colitis and severe extraintestinal complications due to injury of renal endothelial cells, resulting in kidney failure. Since kidney epithelial cells are suggested additional targets for Stxs, we analyzed Madin-Darby canine kidney (MDCK) II epithelial cells for presence of Stx-binding glycosphingolipids (GSLs), determined their distribution to detergent-resistant membranes (DRMs), and ascertained the lipid composition of DRM and non-DRM preparations. Globotriaosylceramide and globotetraosylceramide, known as receptors for Stx1a, Stx2a, and Stx2e, and Forssman GSL as a specific receptor for Stx2e, were found to cooccur with SM and cholesterol in DRMs of MDCK II cells, which was shown using TLC overlay assay detection combined with mass spectrometry. The various lipoforms of GSLs were found to mainly harbor ceramide moieties composed of sphingosine (d18:1) and C24:1/C24:0 or C16:0 FA. The cells were highly refractory toward Stx1a, Stx2a, and Stx2e, most likely due to the absence of Stx-binding GSLs in the apical plasma membrane determined by immunofluorescence confocal laser scanning microscopy. The results suggest that the cellular content of Stx receptor GSLs and their biochemical detection in DRM preparations alone are inadequate to predict cellular sensitivity toward Stxs.


Asunto(s)
Membrana Celular/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Glicoesfingolípidos/metabolismo , Toxina Shiga/metabolismo , Toxina Shiga/toxicidad , Animales , Membrana Celular/efectos de los fármacos , Colesterol/metabolismo , Perros , Riñón/citología , Células de Riñón Canino Madin Darby , Fosfolípidos/metabolismo
6.
J Immunol ; 196(3): 1177-85, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26695372

RESUMEN

Hemolytic uremic syndrome (HUS) is the life-threatenig sequela of intestinal infections by Shiga toxin (Stx)-producing Escherichia coli (STEC) in children. Human neutrophils specifically bind Stx through TLR4, the receptor of LPS. The binding could be considered protective (Stx sequestration) or harmful (toxin delivery to target organs). The amount of Stx on neutrophils is in equilibrium with the amount of Stx present in the gut, and it is also related to renal and neurologic symptoms. The TLR4-mediated interaction of LPS with innate immune cells is hampered by the well-known antibiotic polymyxin B. In this study, we show that the same antibiotic impairs the binding of Stx to neutrophils, also blocking their functional effects (release of CXCL8, formation of neutrophil/platelet aggregates) involved in HUS pathogenesis. Controls for contaminating LPS in Stx-induced neutrophil responses inhibited by polymyxin B were performed. Stx interact with human neutrophils through their A chain, since these leukocytes do not express globotriaosylceramide, the specific receptor for Stx B chains. Consistently, polymyxin B blocked the enzymatic activity of Stx1, Stx2, Stx1 A chain, and the analogous plant protein gelonin, whereas the antibiotic did not show any protective effect on Stx-induced cytotoxicity in globotriaosylceramide-expressing Raji cells. Antibiotic administration is not recommended in human STEC infections during the prodromal intestinal phase, and the toxicity of polymyxin B could further discourage its therapeutic use. However, nontoxic, nonbactericidal polymyxin derivatives have been developed and might be used in animal models of STEC infection to study their efficacy in preventing the onset of HUS during the systemic blood phase of Stx.


Asunto(s)
Antibacterianos/farmacología , Síndrome Hemolítico-Urémico/inmunología , Neutrófilos/efectos de los fármacos , Polimixina B/farmacología , Toxina Shiga/toxicidad , Animales , Citometría de Flujo , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Humanos , Ratones , Neutrófilos/inmunología
7.
Eur J Pediatr ; 177(3): 311-318, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29288280

RESUMEN

Severe complications due to Shigatoxin-associated hemolytic uremic syndrome (STEC-HUS) currently present a serious challenge since no specific treatment for this condition is available. Eculizumab, a terminal complement inhibitor, has been used especially in STEC-HUS patients with severe neurological involvement, but the efficacy remains undetermined. In order to determine its efficacy, we searched the databases Pubmed, Web of Science, Embase, and LiLACS for reports describing outcomes of eculizumab administration in STEC-HUS. We retrieved 11 reports ranging from case reports to cohort studies with the largest study population emanating from the 2011 German outbreak. Outcomes were variable and difficult to interpret in light of the absence of high-quality studies but seemed to point towards potential efficacy of eculizumab if administered early in the course. CONCLUSION: The efficacy of eculizumab in STEC-HUS could not be established nor disproven based on current data, and there is a desperate need for randomized controlled trials. What is known? • Eculizumab has been used in complicated cases of Shigatoxin-associated hemolytic uremic syndrome but the efficacy remains unknown? What is new? • Eculizumab might be efficacious if given early in selected cases of Shigatoxin-associated hemolytic uremic syndrome; however, randomized trials are needed to assess this.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Inactivadores del Complemento/uso terapéutico , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Toxina Shiga/toxicidad , Síndrome Hemolítico-Urémico/etiología , Humanos , Resultado del Tratamiento
8.
Genes Cells ; 21(8): 901-6, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27302278

RESUMEN

Shiga toxin (Stx) is a main virulence factor of Stx-producing Escherichia coli (STEC) that contributes to diarrhea and hemorrhagic colitis and occasionally to fatal systemic complications. Therefore, the development of an antidote to neutralize Stx toxicity is urgently needed. After internalization into cells, Stx is transferred to the Golgi apparatus via a retrograde vesicular transport system. We report here that 2-methylcoprophilinamide (M-COPA), a compound that induces disassembly of the Golgi apparatus by inactivating ADP-ribosylation factor 1 (Arf1), suppresses Stx-induced apoptosis. M-COPA inhibited transport of Stx from the plasma membrane to the Golgi apparatus and suppressed degradation of anti-apoptotic proteins and the activation of caspases. These findings suggest that inhibition of Stx retrograde transport by M-COPA could be a novel approach to suppress Stx toxicity.


Asunto(s)
Factor 1 de Ribosilacion-ADP/genética , Alquenos/farmacología , Antídotos/farmacología , Naftoles/administración & dosificación , Piridinas/administración & dosificación , Toxina Shiga/antagonistas & inhibidores , Escherichia coli Shiga-Toxigénica/efectos de los fármacos , Factor 1 de Ribosilacion-ADP/antagonistas & inhibidores , Alquenos/química , Antídotos/química , Apoptosis/efectos de los fármacos , Apoptosis/genética , Colitis/tratamiento farmacológico , Colitis/microbiología , Diarrea/tratamiento farmacológico , Diarrea/microbiología , Aparato de Golgi/efectos de los fármacos , Humanos , Toxina Shiga/toxicidad , Escherichia coli Shiga-Toxigénica/patogenicidad
9.
J Immunol ; 194(5): 2309-18, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25637016

RESUMEN

Shiga toxin (Stx)-producing Escherichia coli (STEC) cause hemolytic uremic syndrome (HUS). This study investigated whether Stx2 induces hemolysis and whether complement is involved in the hemolytic process. RBCs and/or RBC-derived microvesicles from patients with STEC-HUS (n = 25) were investigated for the presence of C3 and C9 by flow cytometry. Patients exhibited increased C3 deposition on RBCs compared with controls (p < 0.001), as well as high levels of C3- and C9-bearing RBC-derived microvesicles during the acute phase, which decreased after recovery. Stx2 bound to P1 (k) and P2 (k) phenotype RBCs, expressing high levels of the P(k) Ag (globotriaosylceramide), the known Stx receptor. Stx2 induced the release of hemoglobin and lactate dehydrogenase in whole blood, indicating hemolysis. Stx2-induced hemolysis was not demonstrated in the absence of plasma and was inhibited by heat inactivation, as well as by the terminal complement pathway Ab eculizumab, the purinergic P2 receptor antagonist suramin, and EDTA. In the presence of whole blood or plasma/serum, Stx2 induced the release of RBC-derived microvesicles coated with C5b-9, a process that was inhibited by EDTA, in the absence of factor B, and by purinergic P2 receptor antagonists. Thus, complement-coated RBC-derived microvesicles are elevated in HUS patients and induced in vitro by incubation of RBCs with Stx2, which also induced hemolysis. The role of complement in Stx2-mediated hemolysis was demonstrated by its occurrence only in the presence of plasma and its abrogation by heat inactivation, EDTA, and eculizumab. Complement activation on RBCs could play a role in the hemolytic process occurring during STEC-HUS.


Asunto(s)
Vesículas Cubiertas/efectos de los fármacos , Eritrocitos/efectos de los fármacos , Infecciones por Escherichia coli/sangre , Escherichia coli O157/patogenicidad , Síndrome Hemolítico-Urémico/sangre , Toxina Shiga/toxicidad , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/farmacología , Niño , Preescolar , Vesículas Cubiertas/química , Vesículas Cubiertas/inmunología , Activación de Complemento/efectos de los fármacos , Complemento C3/química , Complemento C9/química , Complejo de Ataque a Membrana del Sistema Complemento/química , Ácido Edético/farmacología , Eritrocitos/química , Eritrocitos/inmunología , Eritrocitos/patología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Escherichia coli O157/inmunología , Escherichia coli O157/metabolismo , Femenino , Expresión Génica , Hemólisis/efectos de los fármacos , Síndrome Hemolítico-Urémico/inmunología , Síndrome Hemolítico-Urémico/microbiología , Síndrome Hemolítico-Urémico/patología , Humanos , Lactante , L-Lactato Deshidrogenasa/metabolismo , Masculino , Persona de Mediana Edad , Antagonistas del Receptor Purinérgico P2/farmacología , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/inmunología , Toxina Shiga/química , Toxina Shiga/inmunología , Suramina/farmacología , Trihexosilceramidas/inmunología
10.
Transfus Apher Sci ; 54(2): 203-11, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27156109

RESUMEN

OBJECTIVES: To review the role of complement in glomerular pathologies focusing on thrombotic microangiopathies (TMA) caused by Shiga toxin (Stx) and organ transplantation associated hemolytic uremic syndrome (HUS) as well as C3 glomerulopathy (C3G). METHODS: Examination of literature discussing TMA associated with Stx HUS, transplantation related HUS and C3G. RESULTS: There is an emerging role for complement biology in the renal glomerulus where its inappropriate over-activation is integral to several diseases. Stx HUS patients show evidence of complement activation and the toxin itself can activate complement and inhibit its normal regulation. However, therapeutic complement blockade has not yet proven effective in all circumstances. This may be partly related to late use and a clinical trial could be warranted. Organ transplantation associated HUS has carried a poor prognosis. While case reports supporting the use of complement inhibition exist, there has not been a formal trial. Complement activation in C3G is established but again treatment with complement inhibition has failed to be uniformly beneficial. Here, too, a clinical trial may help determine which subgroup of patients should be treated with these agents. CONCLUSION: Complement plays an important role in the glomerulus but more work is needed to fully understand how it contributes to normal function and pathology. This will help direct appropriate therapy in these diseases.


Asunto(s)
Complemento C3/metabolismo , Glomerulonefritis/sangre , Síndrome Hemolítico-Urémico/sangre , Glomérulos Renales/metabolismo , Trasplante de Riñón/efectos adversos , Toxina Shiga/toxicidad , Microangiopatías Trombóticas/sangre , Glomerulonefritis/etiología , Síndrome Hemolítico-Urémico/etiología , Humanos , Microangiopatías Trombóticas/etiología
11.
Int J Med Microbiol ; 305(1): 20-6, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25465158

RESUMEN

Due to increased release or production of Shiga toxin by Enterohemorrhagic Escherichia coli (EHEC) after exposure to antimicrobial agents, the role of antimicrobial agents in EHEC mediated infections remains controversial. Probiotics are therefore rapidly gaining interest as an alternate therapeutic option. The well-known probiotic strain Escherichia coli Nissle 1917 (EcN) was tested in vitro to determine its probiotic effects on growth, Shiga toxin (Stx) gene expression, Stx amount and associated cytotoxicity on the most important EHEC strains of serotype O104:H4 and O157:H7. Following co-culture of EcN:EHEC in broth for 4 and 24 h, the probiotic effects on EHEC growth, toxin gene expression, Stx amount and cytotoxicity were determined using quantitative real time-PCR, Stx-ELISA and Vero cytotoxicity assays. Probiotic EcN strongly reduced EHEC numbers (cfu) of O104:H4 up to (68%) and O157:H7 to (72.2%) (p<0.05) in LB broth medium whereas the non-probiotic E. coli strain MG1655 had no effect on EHEC growth. The level of stx expression was significantly down-regulated, particularly for the stx2a gene. The stx down-regulation in EcN co-culture was not due to reduced numbers of EHEC. A significant inhibition in Stx amounts and cytotoxicity were also observed in sterile supernatants of EcN:EHEC co-cultures. These findings indicate that probiotic EcN displays strong inhibitory effects on growth, Shiga toxin gene expression, amount and cytotoxicity of EHEC strains. Thus, EcN may be considered as a putative therapeutic candidate, in particular against EHEC O104:H4 and O157:H7.


Asunto(s)
Antibiosis , Escherichia coli/fisiología , Expresión Génica , Probióticos/farmacología , Toxina Shiga/biosíntesis , Toxina Shiga/toxicidad , Animales , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Perfilación de la Expresión Génica , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Toxina Shiga/genética , Células Vero , Factores de Virulencia/biosíntesis , Factores de Virulencia/genética , Factores de Virulencia/toxicidad
12.
Pol Merkur Lekarski ; 39(231): 157-61, 2015 Sep.
Artículo en Polaco | MEDLINE | ID: mdl-26449578

RESUMEN

Toxins produced by the bacteria are of particular interest as potential cargo combat possible for use in a terrorist attack or war. Shiga toxin is usually produced by shiga toxigenic strains of Escherichia coli (STEC - shigatoxigenic Escherichia coli). To infection occurs mostly after eating contaminated beef. Clinical syndromes associated with Shiga toxin diarrhea, hemorrhagic colitis, hemolytic uremic syndrome (HUS - hemolytic uremic syndrome) or thrombotic thrombocytopenic purpura. Treatment is symptomatic. In HUS, in which mortality during an epidemic reaches 20%, extending the kidney injury dialysis may be necessary. Exposure to tetanus toxin produced by Clostridium tetani, resulting in the most generalized tetanus, characterized by increased muscle tension and painful contractions of individual muscle groups. In the treatment beyond symptomatic behavior (among others spasticity medications, anticonvulsants, muscle relaxants) is used tetanus antitoxin and antibiotics (metronidazole choice). A common complication is acute respiratory failure - then it is necessary to implement mechanical ventilation.


Asunto(s)
Armas Biológicas , Bioterrorismo , Toxina Shiga/toxicidad , Toxina Tetánica/toxicidad , Tétanos/inducido químicamente , Animales , Clostridium tetani , Diarrea/inducido químicamente , Diarrea/microbiología , Diarrea/terapia , Escherichia coli , Fluidoterapia , Contaminación de Alimentos , Síndrome Hemolítico-Urémico/inducido químicamente , Síndrome Hemolítico-Urémico/diagnóstico , Síndrome Hemolítico-Urémico/terapia , Humanos , Púrpura Trombocitopénica Trombótica/inducido químicamente , Púrpura Trombocitopénica Trombótica/diagnóstico , Púrpura Trombocitopénica Trombótica/terapia , Tétanos/diagnóstico , Tétanos/terapia
13.
Environ Microbiol ; 16(2): 454-66, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23981100

RESUMEN

Phage-encoded Shiga toxin (Stx) acts as a bacterial defence against the eukaryotic predator Tetrahymena. To function as an effective bacterial anti-predator defence, Stx must kill a broad spectrum of predators. Consistent with that assertion, we show here that bacterially encoded Stx efficiently kills the bacteriovore Acanthamoeba castellanii in co-culture. We also show that, in addition to Stx, the phage-encoded exotoxin, diphtheria toxin (Dtx) expressed by Corynebacterium diphtheriae also can function as part of an anti-predator strategy; it kills Acanthamoeba in co-culture. Interestingly, only exotoxins produced by bacteria internalized by the Acanthamoeba predator are cytolethal; the presence of purified Dtx or Stx in culture medium has no effect on predator viability. This finding is consistent with our results indicating that intoxication of Acanthamoeba by these exotoxins does not require a receptor. Thus bacteria, in the disguise of a food source, function as a 'Trojan Horse', carrying genes encoding an exotoxin into target organisms. This 'Trojan Horse' mechanism of exotoxin delivery into predator cells allows intoxication of predators that lack a cell surface receptor for the particular toxin, allowing bacteria-bearing exotoxins to kill a broader spectrum of predators, increasing the fitness of the otherwise 'defenceless' prey bacteria.


Asunto(s)
Acanthamoeba castellanii/microbiología , Antibiosis , Corynebacterium diphtheriae/fisiología , Toxina Shiga/toxicidad , Acanthamoeba castellanii/metabolismo , Bacteriófagos , Técnicas de Cocultivo , Medios de Cultivo , Toxina Diftérica/toxicidad , Exotoxinas/toxicidad , Fagocitosis , Receptores de Superficie Celular
14.
Environ Microbiol ; 16(4): 963-76, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24034719

RESUMEN

Shiga toxin (Stx)-producing Escherichia coli (STEC) of serogroup O174 are human pathogenic intimin gene (eae)-negative STEC. To facilitate diagnosis and subtyping, we genotypically and phenotypically characterized 25 STEC O174 isolates from humans with different clinical outcomes and from animals and the environment. fliC genotyping resulted in four different genotypes (fliCH2 : n = 5; fliCH8 : n = 8; fliCH21 : n = 11; fliCH46 : n = 1). Twenty-three strains were motile expressing the corresponding H antigen; two non-motile isolates possessed fliCH8 . The stx genotypes and non-stx virulence loci, including toxins, serine-proteases and adhesins correlated well with serotypes but showed no differences with respect to the isolates' origins. Multilocus sequence typing identified seven sequence types that correlated with serotypes. Core gene typing further specified the four serotypes, including a previously unknown O174:H46 combination, and revealed distant relationships of the different serotypes within serogroup O174 and in relation to other haemolytic uremic syndrome (HUS)-associated STEC. Only serotype O174:H21 was associated with HUS. Differences in virulence factors and in the adherence capacity of STEC O174 corroborated this separation into four distinct groups. Our study provides a basis for O174 subtyping, unravels considerable genotypic and phenotypic heterogeneity and sheds light to potential environmental and animal reservoirs.


Asunto(s)
Proteínas de Escherichia coli/genética , Toxina Shiga/genética , Escherichia coli Shiga-Toxigénica/fisiología , Animales , Antibacterianos/farmacología , Adhesión Bacteriana , Bovinos , Línea Celular , Chlorocebus aethiops , Farmacorresistencia Bacteriana Múltiple , Proteínas de Escherichia coli/toxicidad , Genotipo , Humanos , Mucosa Intestinal/microbiología , Datos de Secuencia Molecular , Tipificación de Secuencias Multilocus , Fenotipo , Filogenia , Toxina Shiga/toxicidad , Escherichia coli Shiga-Toxigénica/efectos de los fármacos , Escherichia coli Shiga-Toxigénica/aislamiento & purificación , Células Vero
15.
Toxins (Basel) ; 16(9)2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39330867

RESUMEN

Hemolytic-uremic syndrome (HUS) is a rare complication of an infection with Shiga toxin (Stx)-producing Escherichia coli (STEC-HUS), characterized by severe acute kidney injury, thrombocytopenia and microangiopathic hemolytic anemia, and specific therapy is still lacking. Thrombomodulin (TM) is a multi-domain transmembrane endothelial cell protein and its N-terminal domain has been implicated in the pathophysiology of some cases of HUS. Indeed, the administration of recombinant human TM (rhTM) may have efficacy in HUS. We used a Stx-based murine model of HUS to characterize the role of the N-terminal domain of TM. We show that mice lacking that domain (TMLed (-/-)) are more sensitive to Stx, with enhanced HUS progression seen at 4 days and increased mortality at 7 days post-HUS induction. In spite of these changes, renal function was less affected in surviving Stx-challenged TMLed (-/-) mice compared to their wild-type counterparts TMLed (+/+) at 7 days. Contrary to few clinical case reports from Japan, the administration of rhTM (0.06 mg/kg) to wild-type mice (C57BL/6J) with HUS did not protect against disease progression. This overall promising, but also contradictory body of evidence, requires further systematic preclinical and clinical investigations to clarify the role of TM in HUS as a potential therapeutic strategy.


Asunto(s)
Síndrome Hemolítico-Urémico , Proteínas Recombinantes , Trombomodulina , Trombomodulina/genética , Trombomodulina/uso terapéutico , Animales , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Humanos , Proteínas Recombinantes/uso terapéutico , Proteínas Recombinantes/farmacología , Toxina Shiga/toxicidad , Ratones , Modelos Animales de Enfermedad , Dominios Proteicos
16.
Crit Care Med ; 41(7): 1702-10, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23660733

RESUMEN

OBJECTIVE: In Spring 2011, an unprecedented outbreak of Shiga toxin-producing Escherichia coli serotype O104:H4-associated hemolytic uremic syndrome occurred in Northern Germany. The aim of this study was to describe the clinical characteristics, treatments, and outcomes of critically ill patients with Shiga toxin-producing E. coli-associated hemolytic uremic syndrome during this outbreak. DESIGN, SETTING, AND PATIENTS: Multicenter, retrospective, observational study of critically ill adult patients with Shiga toxin-producing E. coli-associated hemolytic uremic syndrome in six hospitals in Hamburg, Germany, between May 2011 and August 2011. MEASUREMENTS AND MAIN RESULTS: During the study period, 106 patients with Shiga toxin-producing E. coli-associated hemolytic uremic syndrome were admitted to eight ICUs. The median age was 40 years (range, 18-83) with a female:male ratio of 3:1. The median time from onset of clinical symptoms to hospital admission was 3 days and from hospital to ICU admission an additional 3 days. A total of 101 patients (95.3%) had acute renal failure and 78 (73.6%) required renal replacement therapy. Intubation and mechanical ventilation were required in 38 patients (35.8%) and noninvasive ventilation was required in 17 patients (16.0%). The median duration of invasive ventilation was 7 days (range, 1-32 days) and the median ICU stay was 10 days (range, 1-45 days). Fifty-one patients (48.1%) developed sepsis; of these 51 patients, 27 (25.4%) developed septic shock. Seventy patients (66.0%) developed severe neurological symptoms. Ninety-seven patients (91.5%) were treated with plasma exchange and 50 patients (47.2%) received eculizumab (monoclonal anti-C5 antibody). The mortality rate was 4.7%. Mild residual neurological symptoms were present in 21.7% of patients at ICU discharge, and no patient required renal replacement therapy 6 months after ICU admission. CONCLUSIONS: During the 2011 Shiga toxin-producing E. coli-associated hemolytic uremic syndrome outbreak in Germany, critical illness developed rapidly after hospital admission, often in young women. The infection was associated with severe neurological and renal symptoms, requiring mechanical ventilation and renal replacement therapy in a substantial proportion of patients. Overall, recovery was much better than expected.


Asunto(s)
Enfermedad Crítica , Síndrome Hemolítico-Urémico/etiología , Síndrome Hemolítico-Urémico/terapia , Unidades de Cuidados Intensivos , Toxina Shiga/toxicidad , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/epidemiología , Femenino , Síndrome Hemolítico-Urémico/complicaciones , Humanos , Tiempo de Internación , Masculino , Persona de Mediana Edad , Terapia de Reemplazo Renal/métodos , Respiración Artificial/métodos , Estudios Retrospectivos , Sepsis/etiología , Sepsis/terapia , Adulto Joven
18.
Toxicol In Vitro ; 87: 105537, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36535555

RESUMEN

Enterohemorrhagic or Shiga toxin-producing Escherichia coli is a food-poisoning bacterium that grows in the intestine to produce Shiga toxin (Stx). In this study, the effects of 20 polyphenols on the cytotoxicity of Stx1 and Stx2 in Vero cells were investigated. Among these, epigallocatechin gallate, butein, isorhapontigenin, hesperetin, morin, luteolin, resveratrol, and rhapontigenin showed inhibitory effects on the cytotoxicity of Stxs at 0.4 mmol/L. Furthermore, Vero cells pre-treated with these polyphenols were resistant to Stx at 0.4 mmol/L. However, luteolin showed the most potent inhibitory and cytoprotective effect against Stxs at 0.08 mmol/L or more. This inhibitory mechanism of luteolin was determined using a cell-free protein synthesis system and quantitative reverse transcription PCR assay to detect depurination of 28S rRNA in Vero cells. Luteolin did not inhibit the cell-free protein synthesis by Stxs, suggesting that the enzymatic activity of the Stx A subunit was not inhibited by luteolin. The depurination of 28S rRNA by Stxs was also investigated in Vero cells. The 28S rRNA depurination by Stxs was suppressed in Vero cells treated with Stxs which had been pretreated with luteolin. These results suggest that luteolin inhibits the incorporation of Stxs into Vero cells. This is the first report to show that luteolin inhibits the cytotoxicity of both Stx1 and Stx2 by inhibiting the incorporation of Stxs into Vero cells.


Asunto(s)
Toxina Shiga II , Toxina Shiga , Animales , Chlorocebus aethiops , Células Vero , Toxina Shiga/toxicidad , Toxina Shiga I/toxicidad , Toxina Shiga I/metabolismo , Toxina Shiga II/toxicidad , Toxina Shiga II/metabolismo , Luteolina/farmacología , ARN Ribosómico 28S
19.
Nihon Rinsho ; 70(8): 1318-22, 2012 Aug.
Artículo en Japonés | MEDLINE | ID: mdl-22894065

RESUMEN

EHEC is an emergent pathogen which causes haemorrhagic colitis and complications of the potentially fatal HUS. The main virulence factors are the phage-encoded Shiga toxin and the intimate attachment to host cells. Shiga toxin affects cells not only by inhibiting protein biosynthesis but also through the induction of signalling cascades leading to apoptosis. The locus of enterocyte effacement (LEE) encodes a type III secretion system that translocates bacterial effector proteins into the host cell cytoplasm. Intimate attachment is mediated through interactions of the bacterial outer-membrane protein Intimin and the translocated intimin receptor (Tir) on the host cells. Recently, a very rare serogroup strain in humans of E. coli 0104, which carries a stx2a gene in a typical type of EAggEC, appeared in EU and caused a big outbreak of haemorrhagic colitis and HUS.


Asunto(s)
Escherichia coli Enterohemorrágica/patogenicidad , Infecciones por Escherichia coli/microbiología , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/fisiología , Apoptosis/genética , Traslocación Bacteriana/genética , Citocinas/metabolismo , Citoplasma/microbiología , Enterocitos/microbiología , Escherichia coli Enterohemorrágica/clasificación , Escherichia coli Enterohemorrágica/genética , Proteínas de Escherichia coli/fisiología , Síndrome Hemolítico-Urémico/microbiología , Humanos , Biosíntesis de Proteínas/genética , Receptores de Superficie Celular/fisiología , Toxina Shiga/genética , Toxina Shiga/toxicidad , Factores de Virulencia
20.
Nat Chem Biol ; 5(3): 157-65, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19182783

RESUMEN

ADP ribosylation factor 1 (Arf1) plays a critical role in regulating secretory traffic and membrane transport within the Golgi of eukaryotic cells. Arf1 is activated by guanine nucleotide exchange factors (ArfGEFs), which confer spatial and temporal specificity to vesicular transport. We describe here the discovery and characterization of golgicide A, a potent, highly specific, reversible inhibitor of the cis-Golgi ArfGEF GBF1. Inhibition of GBF1 function resulted in rapid dissociation of COPI vesicle coat from Golgi membranes and subsequent disassembly of the Golgi and trans-Golgi network. Secretion of soluble and membrane-associated proteins was arrested at the endoplasmic reticulum-Golgi intermediate compartment, whereas endocytosis and recycling of transferrin were unaffected by GBF1 inhibition. Internalized shiga toxin was arrested within the endocytic compartment and was unable to reach the dispersed trans-Golgi network. Collectively, these results highlight the central role for GBF1 in coordinating bidirectional transport and maintaining structural integrity of the Golgi.


Asunto(s)
Aparato de Golgi/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/fisiología , Piridinas/farmacología , Quinolinas/farmacología , Toxina Shiga/antagonistas & inhibidores , Animales , Chlorocebus aethiops , Proteína Coat de Complejo I/metabolismo , Perros , Endocitosis , Aparato de Golgi/metabolismo , Aparato de Golgi/fisiología , Humanos , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Toxina Shiga/toxicidad , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA