Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
2.
Toxins (Basel) ; 15(12)2023 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-38133194

RESUMEN

Typical hemolytic uremic syndrome (HUS) is mainly caused by Shiga toxin-producing Escherichia coli (STEC) releasing Shiga toxin 2 (Stx2). Two different structures of this AB5 toxin have been described: uncleaved, with intact B and A chains, and cleaved, with intact B and a nicked A chain consisting of two fragments, A1 and A2, connected by a disulfide bond. Despite having the same toxic effect on sensitive cells, the two forms differ in their binding properties for circulating cells, serum components and complement factors, thus contributing to the pathogenesis of HUS differently. The outcome of STEC infections and the development of HUS could be influenced by the relative amounts of uncleaved or cleaved Stx2 circulating in patients' blood. Cleaved Stx2 was identified and quantified for the first time in four out of eight STEC-infected patients' sera by a method based on the inhibition of cell-free translation. Cleaved Stx2 was present in the sera of patients with toxins bound to neutrophils and in two out of three patients developing HUS, suggesting its involvement in HUS pathogenesis, although in association with other bacterial or host factors.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli Shiga-Toxigénica , Humanos , Toxina Shiga II , Toxina Shiga , Neutrófilos , Bacterias , Infecciones por Escherichia coli/microbiología
3.
Microorganisms ; 11(10)2023 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-37894145

RESUMEN

Shiga toxins (Stxs), especially the Stx2a subtype, are the major virulence factors involved in enterohemorrhagic Escherichia coli (EHEC)-associated hemolytic uremic syndrome (eHUS), a life-threatening disease causing acute kidney injury, especially in children. After oral transmission and colonization in the gut, EHEC release Stx. Intracellular cleavage of the Stx A subunit, when followed by reduction, boosts the enzymatic activity that causes damage to targeted cells. This cleavage was assumed to be mostly mediated by furin during Stx intracellular trafficking. To investigate whether this cleavage could occur in the intestine, even prior to entering target cells, Stx2a A subunit structure (intact or cleaved) was characterized after its exposure to specific host factors present in human stool. The molecular weight of Stx2a A subunit/fragments was determined by immunoblotting after electrophoretic separation under reducing conditions. In this study, it was demonstrated that Stx2a is cleaved by certain human stool components. Trypsin and chymotrypsin-like elastase 3B (CELA3B), two serine proteases, were identified as potential candidates that can trigger the extracellular cleavage of Stx2a A subunit directly after its secretion by EHEC in the gut. Whether the observed cleavage indeed translates to natural infections and plays a role in eHUS pathogenesis has yet to be determined. If so, it seems likely that a host's protease profile could affect disease development by changing the toxin's biological features.

4.
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
5.
Biochimie ; 199: 23-35, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35398441

RESUMEN

Among the functions exerted by eukaryotic lactate dehydrogenases, it is of importance the generation of lactate in muscles subjected to fatigue or to limited oxygen availability, with both these conditions triggering a decrease of cellular pH. However, the mutual dependence between lactate dehydrogenase (LDH) catalytic action and lactic acidosis is far from being fully understood. Here we show that the tetrameric LDH from rabbit skeletal muscle undergoes allosteric transitions as a function of pH, i.e. the enzyme obeys Michaelis-Menten kinetics at neutral or slightly alkaline pH values, and features sigmoidal kinetics at pH 6.5 or lower. Remarkably, we also report that a significant dissociation of tetrameric rabbit LDH occurs under acidic conditions, with pyruvate/NAD+ or citrate counteracting this effect. Moreover, citrate strongly activates rabbit LDH, inducing the enzyme to feature Michaelis-Menten kinetics. Further, using primary rabbit skeletal muscle cells we tested the generation of lactate as a function of pH, and we detected a parallel decrease of cytosolic pH and secretion of lactate. Overall, our observations indicate that lactic acidosis is antagonized by LDH dissociation, the occurrence of which is regulated by citrate and by allosteric transitions of the enzyme induced by pyruvate.


Asunto(s)
Acidosis Láctica , L-Lactato Deshidrogenasa , Animales , Citratos , Concentración de Iones de Hidrógeno , Cinética , L-Lactato Deshidrogenasa/metabolismo , Ácido Láctico , Músculo Esquelético/metabolismo , Ácido Pirúvico , Conejos
6.
Biosci Rep ; 42(1)2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35048959

RESUMEN

The aerobic energetic metabolism of eukaryotic cells relies on the glycolytic generation of pyruvate, which is subsequently channelled to the oxidative phosphorylation taking place in mitochondria. However, under conditions limiting oxidative phosphorylation, pyruvate is coupled to alternative energetic pathways, e.g. its reduction to lactate catalyzed by lactate dehydrogenases (LDHs). This biochemical process is known to induce a significant decrease in cytosolic pH, and is accordingly denoted lactic acidosis. Nevertheless, the mutual dependence of LDHs action and lactic acidosis is far from being fully understood. Using human LDH-A, here we show that when exposed to acidic pH this enzyme is subjected to homotropic allosteric transitions triggered by pyruvate. Conversely, human LDH-A features Michaelis-Menten kinetics at pH values equal to 7.0 or higher. Further, citrate, isocitrate, and malate were observed to activate human LDH-A, both at pH 5.0 and 6.5, with citrate and isocitrate being responsible for major effects. Dynamic light scattering (DLS) experiments revealed that the occurrence of allosteric kinetics in human LDH-A is mirrored by a consistent dissociation of the enzyme tetramer, suggesting that pyruvate promotes tetramer association under acidic conditions. Finally, using the human liver cancer cell line HepG2 we isolated cells featuring cytosolic pH equal to 7.3 or 6.5, and we observed a concomitant decrease in cytosolic pH and lactate secretion. Overall, our observations indicate the occurrence of a negative feedback between lactic acidosis and human LDH-A activity, and a complex regulation of this feedback by pyruvate and by some intermediates of the Krebs cycle.


Asunto(s)
Lactato Deshidrogenasa 5/química , Piruvatos/química , Humanos , Concentración de Iones de Hidrógeno , Ácido Láctico
7.
J Pediatr ; 237: 34-40.e1, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34197890

RESUMEN

OBJECTIVE: To analyze the results of an enhanced laboratory-surveillance protocol for bloody diarrhea aimed at identifying children with Shiga toxin-producing Escherichia coli (STEC) infection early in the course of the disease toward the early identification and management of patients with hemolytic uremic syndrome (HUS). STUDY DESIGN: The study (2010-2019) involved a referral population of 2.3 million children. Stool samples of patients with bloody diarrhea were screened for Shiga toxin (Stx) genes. Positive patients were rehydrated and monitored for hemoglobinuria until diarrhea resolved or STEC-HUS was diagnosed. RESULTS: A total of 4767 children were screened; 214 (4.5%) were positive for either Stx1 (29.0%) or Stx2 (45.3%) or both Stx1+2 (25.7%); 34 patients (15.9%) developed STEC-HUS (0.71% of bloody diarrheas). Hemoglobinuria was present in all patients with HUS. Patients with Stx2 alone showed a greater risk of STEC-HUS (23.7% vs 12.7%) and none of the patients with Stx1 alone developed HUS. During the same period of time, 95 other patients were diagnosed STEC-HUS but were not captured by the screening program (26 had nonbloody diarrhea, 11 came from areas not covered by the screening program, and 58 had not been referred to the screening program, although they did meet the inclusion criteria). At HUS presentation, serum creatinine of patients identified by screening was significantly lower compared with that of the remaining patients (median 0.9 vs 1.51 mg/dL). CONCLUSIONS: Nearly 1% of children with bloody diarrhea developed STEC-HUS, and its diagnosis was anticipated by the screening program for Stx. The screening of bloody diarrhea for Stx is recommended, and monitoring patients carrying Stx2 with urine dipstick for hemoglobinuria is suggested to identify the renal complication as early as possible.


Asunto(s)
Diarrea/microbiología , Infecciones por Escherichia coli/diagnóstico , Hemorragia Gastrointestinal/microbiología , Síndrome Hemolítico-Urémico/microbiología , Tamizaje Masivo/métodos , Escherichia coli Shiga-Toxigénica/aislamiento & purificación , Adolescente , Niño , Preescolar , Diagnóstico Precoz , Infecciones por Escherichia coli/complicaciones , Femenino , Hemorragia Gastrointestinal/diagnóstico , Genes Bacterianos , Síndrome Hemolítico-Urémico/diagnóstico , Síndrome Hemolítico-Urémico/epidemiología , Síndrome Hemolítico-Urémico/terapia , Humanos , Lactante , Recién Nacido , Italia , Masculino , Toxinas Shiga/genética , Escherichia coli Shiga-Toxigénica/genética , Resultado del Tratamiento , Adulto Joven
8.
Am J Pathol ; 191(5): 795-804, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33652019

RESUMEN

This review focuses on typical hemolytic uremic syndrome (HUS), a life-threatening sequela of human infections caused, particularly in children, by Shiga toxin-producing Escherichia coli strains. Thrombotic microangiopathy of the brain and the kidney is the end point of toxin action, resulting in the hallmarks of HUS (ie, thrombocytopenia, anemia, and acute renal failure). A growing body of evidence points to the role of extracellular vesicles released in the blood of patients by toxin-challenged circulating cells (monocytes, neutrophils, and erythrocytes) and platelets, as a key factor in the pathogenesis of HUS. This review provides i) an updated description of the pathogenesis of Shiga toxin-producing E. coli infections; ii) an analysis of blood cell-derived extracellular vesicles, and of their parent cells, as triggering factors in HUS; and iii) a model explaining why Shiga toxin-containing vesicles dock preferentially to the endothelia of target organs.


Asunto(s)
Infecciones por Escherichia coli/patología , Síndrome Hemolítico-Urémico/patología , Escherichia coli Shiga-Toxigénica/fisiología , Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Anemia/etiología , Anemia/patología , Células Endoteliales/patología , Eritrocitos/patología , Vesículas Extracelulares/patología , Síndrome Hemolítico-Urémico/complicaciones , Humanos , Monocitos/patología , Neutrófilos/patología , Trombocitopenia/etiología , Trombocitopenia/patología
9.
Toxins (Basel) ; 13(2)2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33530614

RESUMEN

The pathogenesis of Escherichia coli-induced hemolytic uremic syndrome (eHUS) caused by infections with pathogenic Shiga toxin (Stx) producing E. coli (STEC) is centered on bacterial (e.g., Stx) and host factors (circulating cells, complement system, serum proteins) whose interaction is crucial for the immediate outcome and for the development of this life-threatening sequela. Stx2a, associated to circulating cells (early toxemia) or extracellular vesicles (late toxemia) in blood, is considered the main pathogenic factor in the development of eHUS. Recently, it was found that the functional properties of Stx2a (binding to circulating cells and complement components) change according to modifications of the structure of the toxin, i.e., after a single cleavage of the A subunit resulting in two fragments, A1 and A2, linked by a disulfide bridge. Herein, we describe a method to be used for the detection of the cleaved form of Stx2a in the serum of STEC-infected or eHUS patients. The method is based on the detection of the boosted inhibitory activity of the cleaved toxin, upon treatment with reducing agents, on a rabbit cell-free translation system reconstituted with human ribosomes. The method overcomes the technical problem caused by the presence of inhibitors of translation in human serum that have been stalled by the addition of RNAase blockers and by treatment with immobilized protein G. This method, allowing the detection of Stx2a at concentrations similar to those found by ELISA in the blood of STEC-infected patients, could be a useful tool to study the contribution of the cleaved form of Stx2a in the pathogenesis of eHUS.


Asunto(s)
Bioensayo , Infecciones por Escherichia coli/diagnóstico , Síndrome Hemolítico-Urémico/diagnóstico , Toxina Shiga II/sangre , Escherichia coli Shiga-Toxigénica/metabolismo , Animales , Biomarcadores/sangre , Sistema Libre de Células/metabolismo , Infecciones por Escherichia coli/sangre , Infecciones por Escherichia coli/microbiología , Síndrome Hemolítico-Urémico/sangre , Síndrome Hemolítico-Urémico/microbiología , Humanos , Valor Predictivo de las Pruebas , Conejos , Reticulocitos/metabolismo , Ribosomas/metabolismo
10.
Pediatr Nephrol ; 35(10): 1997-2001, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32734345

RESUMEN

BACKGROUND: Shigatoxin (Stx)-producing Escherichia coli (STEC) are the most common causes of hemolytic uremic syndrome (STEC-HUS). The aim of our study is to compare the risk of developing STEC-HUS in relation to the type of Stx genes (Stx1, Stx2, or both). METHODS: This is a prospective, observational, multicenter study involving 63 pediatric units in Northern Italy (ItalKid-HUS Network). STEC-infected children were identified within a screening program for bloody diarrhea during a 10-year period (2010-2019). Stx genes were detected by reverse dot blot or real-time PCR. After the identification of STEC infection, children were followed until diarrhea complete recovery for the possible development of STEC-HUS. RESULTS: Of the 214 Stx-positive patients, 34 (15.9%) developed STEC-HUS. The risk of HUS in STEC-infected children with Stx1 (n: 62; 29.0%) and Stx2 (n: 97; 45.3%) was respectively 0% and 23.7%, while in patients carrying both Stx1 and Stx2 (n: 55; 25.7%), the risk was 12.7% (p: 0.001). CONCLUSIONS: Our data confirm that Stx1 is a very rare cause of STEC-HUS and demonstrate that the risk of STEC-HUS halves in the case of Stx1+2-producing Escherichia coli infection compared with infections where Stx2 is present alone. This observation is helpful in assessing the risk of individual STEC-infected patients for the development of HUS and suggests that Stx1, in the presence of Stx2, might exert a protective role possibly by receptor competition.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Síndrome Hemolítico-Urémico/epidemiología , Toxina Shiga I/toxicidad , Toxina Shiga II/toxicidad , Escherichia coli Shiga-Toxigénica/genética , Niño , Preescolar , Infecciones por Escherichia coli/complicaciones , Femenino , Síndrome Hemolítico-Urémico/microbiología , Humanos , Lactante , Tipificación Molecular , Estudios Prospectivos , Factores Protectores , Medición de Riesgo , Toxina Shiga I/genética , Toxina Shiga I/aislamiento & purificación , Toxina Shiga II/genética , Toxina Shiga II/aislamiento & purificación , Escherichia coli Shiga-Toxigénica/aislamiento & purificación
11.
Thromb Haemost ; 120(1): 107-120, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31858520

RESUMEN

Hemolytic uremic syndrome (HUS), the leading cause of acute renal failure in children (< 3 years), is mainly related to Shiga toxins (Stx)-producing Escherichia coli (STEC) infections. STEC are confined to the gut resulting in hemorrhagic colitis, whereas Stx are delivered in blood to target kidney and brain, with unclear mechanisms, triggering HUS in 5 to 15% of infected children. Stx were found on circulating cells, free in sera (soluble Stx) or in blood cell-derived microvesicles (particulate Stx), whereby the relationship between these forms of circulating toxins is unclear. Here, we have examined 2,846 children with bloody diarrhea and found evidence of STEC infection in 5%. Twenty patients were enrolled to study the natural course of STEC infections before the onset of HUS. In patients, Stx were found to be associated to circulating cells and/or free and functionally active in sera. In most children, Stx were bound to neutrophils when high amounts of toxins were found in feces. Time-course analysis showed that Stx increased transiently in patients' sera while the decrease of toxin amount on leukocytes was observed. Notably, patients who recovered (85%) displayed different settings than those who developed HUS (15%). The distinctive feature of the latter group was the presence in blood of particulate Stx2 (Stx2 sedimented at g-forces corresponding to 1 µm microvesicles) the day before diagnosis of HUS, during the release phase of toxins from circulating cells. This observation strongly suggests the involvement of blood cell-derived particulate Stx2 in the transition from hemorrhagic colitis to HUS.


Asunto(s)
Infecciones por Escherichia coli/metabolismo , Síndrome Hemolítico-Urémico/metabolismo , Riñón/metabolismo , Neutrófilos/metabolismo , Material Particulado/sangre , Toxina Shiga II/sangre , Escherichia coli Shiga-Toxigénica/fisiología , Adolescente , Línea Celular , Niño , Preescolar , ADN Bacteriano/genética , Heces/microbiología , Femenino , Humanos , Lactante , Recién Nacido , Riñón/patología , Masculino , Toxina Shiga II/genética
12.
Oxid Med Cell Longev ; 2019: 6528106, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31396304

RESUMEN

In the cold environments of the interstellar medium, a variety of molecules in which a hydrogen (H) atom has been replaced by its heavier isotope deuterium (D) can be found. From its emergence, life had to counteract the toxic action of many agents, which posed a constant threat to its development and propagation. Oxygen-reactive species are archaic toxicants that lead to protein damage and genomic instability. Most of the oxidative lesions involve cleavage of C-H bonds and H abstraction. According to free radical chemistry principles, the substitution of D for H in oxidation-sensitive positions of cellular components should confer protection against the oxidative attack without compromising the chemical identity of the compounds. Here, we show that deuterated nucleosides and proteins protect from oxidative damage. Our data suggest a new, subtle but likely role of D in terrestrial life's evolution in that its inclusion in critical biomolecules might have facilitated their resistance during the infinite generations of life entities, cells, and organisms.


Asunto(s)
Deuterio/química , Estrés Oxidativo , Supervivencia Celular/efectos de los fármacos , Sistema Libre de Células , Daño del ADN/efectos de los fármacos , Radicales Libres/química , Productos Finales de Glicación Avanzada/análisis , Humanos , Células Jurkat , Nucleósidos/química , Nucleósidos/metabolismo , Nucleósidos/farmacología , Estrés Oxidativo/efectos de los fármacos , Carbonilación Proteica , Proteínas/química , Proteínas/metabolismo
13.
Sci Rep ; 9(1): 7199, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-31076590

RESUMEN

Proteasome inhibition represents an important anticancer strategy. Here, we studied the mechanisms at the basis of the pro-apoptotic activity of the standardized decoction of Hemidesmus indicus, a plant evoking a complex anticancer activity, and explored its inhibition of proteasome activity in human leukemia cells. Additionally, we preliminary tested the cytotoxicity of some H. indicus's phytochemicals on leukemia cells and their intestinal absorption on a human intestinal epithelium model consisting of a monolayer of differentiated Caco2 cells. We observed a potent antileukemic effect for H. indicus, imputable to the modulation of different critical targets at protein and mRNA levels and the reduction of the 26S proteasome expression. We found that some phytomarkers of H. indicus decoction passed through the enterocyte monolayer. Overall, our study supports the pharmacological potential of H. indicus, which can represent an interesting botanical drug in the oncological area.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Hemidesmus/química , Inhibidores de Proteasoma/farmacología , Especies Reactivas de Oxígeno/metabolismo , Antineoplásicos Fitogénicos/química , Células CACO-2 , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Absorción Intestinal , Células Jurkat , MicroARNs/genética , Extractos Vegetales/química , Extractos Vegetales/farmacología , Raíces de Plantas/química , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/química
14.
Cell Microbiol ; 21(5): e13000, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30578712

RESUMEN

Hemolytic uremic syndrome (eHUS) is a severe complication of human infections with Shiga toxins (Stxs)-producing Escherichia coli. A key step in the pathogenesis of eHUS is the interaction of Stxs with blood components before the targeting of renal endothelial cells. Here, we show that a single proteolytic cleavage in the Stx2a A-subunit, resulting into two fragments (A1 and A2) linked by a disulfide bridge (cleaved Stx2a), dictates different binding abilities. Uncleaved Stx2a was confirmed to bind to human neutrophils and to trigger leukocyte/platelet aggregate formation, whereas cleaved Stx2a was ineffective. Conversely, binding of complement factor H was confirmed for cleaved Stx2a and not for uncleaved Stx2a. It is worth noting that uncleaved and cleaved Stx2a showed no differences in cytotoxicity for Vero cells or Raji cells, structural conformation, and contaminating endotoxin. These results have been obtained by comparing two Stx2a batches, purified in different laboratories by using different protocols, termed Stx2a(cl; cleaved toxin, Innsbruck) and Stx2a(uncl; uncleaved toxin, Bologna). Stx2a(uncl) behaved as Stx2a(cl) after mild trypsin treatment. In this light, previous controversial results obtained with purified Stx2a has to be critically re-evaluated; furthermore, characterisation of the structure of circulating Stx2a is mandatory to understand eHUS-pathogenesis and to develop therapeutic approaches.


Asunto(s)
Escherichia coli/química , Toxina Shiga II/química , Toxina Shiga II/metabolismo , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Chlorocebus aethiops , Dicroismo Circular , Factor H de Complemento/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Fluorescencia , Humanos , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Unión Proteica , Conformación Proteica , Toxina Shiga II/genética , Trihexosilceramidas/metabolismo , Tripsina , Células Vero
15.
Toxins (Basel) ; 10(9)2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30231570

RESUMEN

Shiga toxin 2a (Stx2a) is the main virulence factor produced by pathogenic Escherichia coli strains (Stx-producing E. coli, STEC) responsible for hemorrhagic colitis and the life-threatening sequela hemolytic uremic syndrome in children. The toxin released in the intestine by STEC targets the globotriaosylceramide receptor (Gb3Cer) present on the endothelial cells of the brain and the kidney after a transient blood phase during which Stx2a interacts with blood components, such as neutrophils, which, conversely, recognize Stx through Toll-like receptor 4 (TLR4). Among non-cellular blood constituents, human amyloid P component (HuSAP) is considered a negative modulating factor that specifically binds Stx2a and impairs its toxic action. Here, we show that the soluble extracellular domain of TLR4 inhibits the binding of Stx2a to neutrophils, assessed by indirect flow cytometric analysis. Moreover, by using human sensitive Gb3Cer-expressing cells (Raji cells) we found that the complex Stx2a/soluble TLR4 escaped from capture by HuSAP allowing the toxin to target and damage human cells, as assayed by measuring translation inhibition, the typical Stx-induced functional impairment. Thus, soluble TLR4 stood out as a positive modulating factor for Stx2a. In the paper, these findings have been discussed in the context of the pathogenesis of hemolytic uremic syndrome.


Asunto(s)
Componente Amiloide P Sérico/metabolismo , Toxina Shiga II/toxicidad , Receptor Toll-Like 4/metabolismo , Línea Celular Tumoral , Humanos , Neutrófilos/metabolismo , Dominios Proteicos
16.
Int J Med Microbiol ; 308(7): 940-946, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29983334

RESUMEN

The life-threatening sequela of hemorrhagic colitis induced by Shiga toxins (Stx)-producing Escherichia coli (STEC) infections in humans is hemolytic uremic syndrome (HUS), the main cause of acute renal failure in early childhood. The key step in the pathogenesis of HUS is the appearance of Stx in the blood of infected patients because these powerful virulence factors are capable of inducing severe microangiopathic lesions in the kidney. During precocious toxemia, which occurs in patients before the onset of HUS during the intestinal phase, Stx bind to several different circulating cells. An early response of these cells might include the release of proinflammatory mediators associated with the development of HUS. Here, we show that primary human monocytes stimulated with Shiga toxin 1a (Stx1a) through the glycolipid receptor globotriaosylceramide released larger amounts of proinflammatory molecules (IL-1ß, TNFα, IL-6, G-CSF, CXCL8, CCL2, CCL4) than Stx1a-treated neutrophils. The mediators (except IL-1ß) are among the top six proinflammatory mediators found in the sera from patients with HUS in different studies. The molecules appear to be involved in different pathogenetic steps of HUS, i.e. sensitization of renal endothelial cells to the toxin actions (IL-1ß, TNFα), activation of circulating monocytes and neutrophils (CXCL8, CCL2, CCL4) and increase in neutrophil counts in patients with poor prognosis (G-CSF). Hence, a role of circulating monocytes in the very early phases of the pathogenetic process culminating with HUS can be envisaged. Impairment of the events of precocious toxemia would prevent or reduce the risk of HUS in STEC-infected children.


Asunto(s)
Citocinas/sangre , Síndrome Hemolítico-Urémico/patología , Monocitos/metabolismo , Toxina Shiga I/metabolismo , Escherichia coli Shiga-Toxigénica/patogenicidad , Trihexosilceramidas/metabolismo , Células Cultivadas , Citocinas/metabolismo , Síndrome Hemolítico-Urémico/microbiología , Humanos , Interleucina-8/sangre , Neutrófilos/metabolismo
17.
Toxins (Basel) ; 10(2)2018 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-29385045

RESUMEN

Shiga toxins (Stx) released by Stx-producing E. coli (STEC) are virulence factors that are most closely 3associated with hemolytic uremic syndrome (HUS), a life-threatening complication of intestinal infections by STEC. Stx have to enter into the circulatory system before they are delivered to target organs and cause damage. The presence of Stx in sera could be a risk indicator for HUS development. However, the detection of Stx, particularly Stx2, has been difficult due to the presence of Stx2-binding components in human serum. Here, we report new ELISA-based methods for the detection of Stx1 and Stx2 in human serum and the effect of guanidinium chloride on enhancing the sensitivity for the detection of Stx2. The recovery rate for Stx2 was 62% when Stx2-spiked serum samples were treated with guanidinium chloride at a concentration of 200 mM, in contrast to 17% without guanidinium chloride treatment. The effectiveness of guanidinium chloride treatment for the detection of Stx2 in human serum was validated using sera from STEC-infected patients. Coimmunoprecipitation results indicated a specific physical interaction between Stx2 and the human serum amyloid P component (HuSAP) in human serum samples. Our in vitro study demonstrated that the inhibition from HuSAP alone for the detection of Stx2 was only 20%, much less than 69.6% from human serum at Stx2 level 10 ng/mL, suggesting that there may be other factors that bind Stx2 in human serum. This study indicates that treatment of serum samples with guanidinium chloride may be useful for the early and sensitive detection of Stx2 in sera of STEC-infected patients, so preventive measures can be adopted in a timely manner.


Asunto(s)
Infecciones por Escherichia coli/diagnóstico , Toxina Shiga II/sangre , Escherichia coli Shiga-Toxigénica , Ensayo de Inmunoadsorción Enzimática , Infecciones por Escherichia coli/sangre , Guanidina/química , Humanos , Componente Amiloide P Sérico/química , Toxina Shiga I/sangre , Toxina Shiga II/química
18.
Biochem Biophys Res Commun ; 485(4): 742-745, 2017 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-28257841

RESUMEN

Most cancer cells use aerobic glycolysis to fuel their growth and many efforts are made to selectively block this metabolic pathway in cancer cells by inhibiting lactate dehydrogenase A (LDHA). However, LDHA is a moonlighting protein which exerts functions also in the nucleus as a factor associated to transcriptional complexes. Here we found that two small molecules which inhibit the enzymatic activity of LDHA hinder the transcription of histone 2B gene independently from the block of aerobic glycolysis. Moreover, we observed that silencing this gene reduces cell replication, hence suggesting that the inhibition of LDHA can also affect the proliferation of normal non-glycolysing dividing cells.


Asunto(s)
Glucólisis/genética , Histonas/genética , L-Lactato Deshidrogenasa/genética , Transcripción Genética/genética , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Galactosa/farmacología , Glucosa/farmacología , Glucólisis/efectos de los fármacos , Células HCT116 , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/antagonistas & inhibidores , L-Lactato Deshidrogenasa/metabolismo , Lactato Deshidrogenasa 5 , Ácido Oxámico/farmacología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bibliotecas de Moléculas Pequeñas/farmacología , Transcripción Genética/efectos de los fármacos
19.
Biochem Biophys Res Commun ; 483(3): 936-940, 2017 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-28082201

RESUMEN

Protein synthesis is one of the main cellular functions inhibited during hypertonic challenge. The subsequent accumulation of the compatible osmolyte betaine during the later adaptive response allows not only recovery of translation but also its stimulation. In this paper, we show that betaine modulates translation by enhancing the formation of cap-independent 48 S pre-initiation complexes, leaving cap-dependent 48 S pre-initiation complexes basically unchanged. In the presence of betaine, CrPV IRES- and sodium-dependent neutral amino acid transporter-2 (SNAT2) 5'-UTR-driven translation is 2- and 1.5-fold stimulated in MCF7 cells, respectively. Thus, betaine could provide an advantage in translation of messengers coding for proteins implicated in the response of cells to different stressors, which are often recognized by ribosomal 40 S subunit through simplified cap-independent mechanisms.


Asunto(s)
Betaína/metabolismo , Betaína/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Caperuzas de ARN/metabolismo , Regiones no Traducidas 5' , Sistema de Transporte de Aminoácidos A/metabolismo , Animales , Sistema Libre de Células , Humanos , Soluciones Hipertónicas , Luciferasas/genética , Luciferasas/metabolismo , Células MCF-7 , Presión Osmótica , Polirribosomas/metabolismo , Biosíntesis de Proteínas/genética , Conejos , Reticulocitos/efectos de los fármacos , Reticulocitos/metabolismo
20.
Nat Protoc ; 11(7): 1309-25, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27336708

RESUMEN

We describe a cell-free translation system for evaluating the activity of ribosomes stringently purified from human cells. This system is based on in vitro reconstitution of the cellular translation machinery, in which a ribosome-free rabbit reticulocyte lysate (RRL) is reassembled with human ribosomes and in vitro-transcribed reporter mRNAs. The protocol describes the preparation of the RRL-derived fractions, purification of ribosomes devoid of detectable nonribosomal-associated factors, and assembly of the reactions to evaluate ribosomal translational efficiency and fidelity using appropriate reporter transcripts. The whole procedure can be completed in ∼2.5 d (plus 2 weeks for RRL preparation and cell expansion time). This protocol can be applied to study intrinsic functional properties (cis-acting element-mediated translation initiation or translational fidelity) of ribosome populations from different sources (including nonhuman origin). It is therefore useful for the characterization of ribosomal function in ribosomopathies and cancer, and it will be applicable in the emerging fields of ribosome diversity and specialized ribosomes.


Asunto(s)
Fraccionamiento Celular/métodos , Sistema Libre de Células/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/genética , Ribosomas/genética , Transcripción Genética , Animales , Genes Reporteros , Humanos , Conejos , Reticulocitos/metabolismo , Ribosomas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA