Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Am J Pathol ; 180(3): 1040-1048, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22203055

RESUMEN

Shiga toxin (Stx)-producing Escherichia coli is the main etiological agent that causes hemolytic uremic syndrome (HUS), a microangiopathic disease characterized by hemolytic anemia, thrombocytopenia, and acute renal failure. Although direct cytotoxic effects on endothelial cells by Stx are the primary pathogenic event, there is evidence that indicates the inflammatory response mediated by polymorphonuclear neutrophils and monocytes as the key event during HUS development. Because the chemokine receptor CCR1 participates in the pathogenesis of several renal diseases by orchestrating myeloid cell kidney infiltration, we specifically addressed the contribution of CCR1 in a murine model of HUS. We showed that Stx type 2-treated CCR1(-/-) mice have an increased survival rate associated with less functional and histological renal damage compared with control mice. Stx type 2-triggered neutrophilia and monocytosis and polymorphonuclear neutrophil and monocyte renal infiltration were significantly reduced and delayed in CCR1(-/-) mice compared with control mice. In addition, the increase of the inflammatory cytokines (tumor necrosis factor-α and IL-6) in plasma was delayed in CCR1(-/-) mice compared with control mice. These data demonstrate that CCR1 participates in cell recruitment to the kidney and amplification of the inflammatory response that contributes to HUS development. Blockade of CCR1 could be important to the design of future therapies to restrain the inflammatory response involved in the development of HUS.


Asunto(s)
Síndrome Hemolítico-Urémico/inducido químicamente , Receptores CCR1/fisiología , Toxina Shiga II/toxicidad , Animales , Médula Ósea/patología , Creatina/metabolismo , Síndrome Hemolítico-Urémico/patología , Interleucina-6/metabolismo , Túbulos Renales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Células Mieloides/fisiología , Receptores CCR1/deficiencia , Tasa de Supervivencia , Factor de Necrosis Tumoral alfa/metabolismo , Urea/metabolismo
2.
J Clin Immunol ; 32(3): 622-31, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22228570

RESUMEN

PURPOSE: The interaction of Shiga toxin (Stx) and/or lipopolysaccharide (LPS) with monocytes (Mo) may be central to the pathogenesis of hemolytic uremic syndrome (HUS), providing the cytokines necessary to sensitize endothelial cells to Stx action. We have previously demonstrated phenotypical alterations in Mo from HUS patients, including increased number of CD16+ Mo. Our aim was to investigate cytokine production in Mo from HUS patients. METHODS: We evaluated TNF-α and IL-10 intracellular contents and secretion in the different Mo subsets in mild (HUS 1) and moderate/severe (HUS 2 + 3) patients. As controls, we studied healthy (HC) and infected children (IC). We also studied Mo responsive capacity towards LPS, measuring the modulation of Mo surface molecules and cytokine production. RESULTS: In basal conditions, the intracellular measurement of TNF-α and IL-10 revealed that the highest number of cytokine-producing Mo was found in HUS 2 + 3 and IC, whereas LPS caused a similar increase in TNF-α and IL-10-producing Mo for all groups. However, when evaluating the release of TNF-α and IL-10, we found a diminished secretion capacity in the entire HUS group and IC compared to HC in basal and LPS conditions. Similarly, a lower Mo response to LPS in HUS 2 + 3 and IC groups was observed when surface markers were studied. CONCLUSION: These results indicate that Mo from severe cases of HUS, similar to IC but different to mild HUS cases, present functional changes in Mo subpopulations and abnormal responses to LPS.


Asunto(s)
Síndrome Hemolítico-Urémico/inmunología , Interleucina-10/inmunología , Monocitos/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Niño , Preescolar , Femenino , Humanos , Lactante , Interleucina-10/sangre , Lipopolisacáridos/inmunología , Masculino , Factor de Necrosis Tumoral alfa/sangre
3.
Immunology ; 129(4): 600-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20102414

RESUMEN

The membrane-anchored form of the chemokine fractalkine (CX(3)CL1) has been identified as a novel adhesion molecule that interacts with its specific receptor (CX(3)CR1) expressed in monocytes, T cells and natural killer cells to induce adhesion. In addition, CX(3)CL1 can be cleaved from the cell membrane to induce chemotaxis of CX(3)CR1-expressing leucocytes. Recently, marked variations in CX(3)CR1 monocyte expression have been observed during several pathological conditions. Regulation of CX(3)CR1 in monocytes during basal or inflammatory/anti-inflammatory conditions is poorly understood. The aim of this study was therefore to examine CX(3)CR1 expression during monocyte maturation and the effect of soluble mediators on this process. We found that basal expression of CX(3)CR1 in fresh monocytes was reduced during culture, and that lipopolysacchairde accelerated this effect. In contrast, interleukin-10 and interferon-gamma treatment abrogated CX(3)CR1 down-modulation, through a phosphatidylinositol 3 kinase-dependent pathway. Most importantly, CX(3)CR1 membrane expression correlated with monocyte CX(3)CL1-dependent function. Taken together, our data demonstrate that CX(3)CR1 expression in monocytes can be modulated, and suggest that alterations in their environment are able to influence CX(3)CL1-dependent functions, such as chemotaxis and adhesion, leading to changes in the kinetics, composition and/or functional status of the leucocyte infiltrate.


Asunto(s)
Interferón gamma/farmacología , Interleucina-10/farmacología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Quimiocina/biosíntesis , Receptor 1 de Quimiocinas CX3C , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Interferón gamma/inmunología , Interleucina-10/inmunología , Monocitos/inmunología , Receptores de Quimiocina/inmunología , Factores de Tiempo
4.
Toxins (Basel) ; 9(11)2017 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-29068360

RESUMEN

Shiga toxin (Stx), produced by Escherichia coli, is the main pathogenic factor of diarrhea-associated hemolytic uremic syndrome (HUS), which is characterized by the obstruction of renal microvasculature by platelet-fibrin thrombi. It is well known that the oxidative imbalance generated by Stx induces platelet activation, contributing to thrombus formation. Moreover, activated platelets release soluble CD40 ligand (sCD40L), which in turn contributes to oxidative imbalance, triggering the release of reactive oxidative species (ROS) on various cellular types. The aim of this work was to determine if the interaction between the oxidative response and platelet-derived sCD40L, as consequence of Stx-induced endothelium damage, participates in the pathogenic mechanism during HUS. Activated human glomerular endothelial cells (HGEC) by Stx2 induced platelets to adhere to them. Although platelet adhesion did not contribute to endothelial damage, high levels of sCD40L were released to the medium. The release of sCD40L by activated platelets was inhibited by antioxidant treatment. Furthermore, we found increased levels of sCD40L in plasma from HUS patients, which were also able to trigger the respiratory burst in monocytes in a sCD40L-dependent manner. Thus, we concluded that platelet-derived sCD40L and the oxidative response are reciprocally stimulated during Stx2-associated HUS. This process may contribute to the evolution of glomerular occlusion and the microangiopathic lesions.


Asunto(s)
Ligando de CD40/sangre , Células Endoteliales/efectos de los fármacos , Síndrome Hemolítico-Urémico/sangre , Toxina Shiga/toxicidad , Células Cultivadas , Niño , Preescolar , Células Endoteliales/patología , Femenino , Síndrome Hemolítico-Urémico/inducido químicamente , Humanos , Lactante , Riñón/metabolismo , Riñón/patología , Masculino , Microvasos , Monocitos/metabolismo , Estrés Oxidativo , Activación Plaquetaria/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
5.
J Leukoc Biol ; 78(4): 853-61, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16046554

RESUMEN

Monocytes (Mo) mediate central functions in inflammation and immunity. Different subpopulations of Mo with distinct phenotype and functional properties have been described. Here, we investigate the phenotype and function of peripheral Mo from children with hemolytic uremic syndrome (HUS). For this purpose, blood samples from patients in the acute period of HUS (HUS AP) were obtained on admission before dialysis and/or transfusion. The Mo phenotypic characterization was performed on whole blood by flow cytometry, and markers associated to biological functions were selected: CD14 accounting for lipopolysaccharide (LPS) responsiveness, CD11b for adhesion, Fc receptor for immunoglobulin G type I (FcgammaRI)/CD64 for phagocytosis and cytotoxicity, and human leukocyte antigen (HLA)-DR for antigen presentation. Some of these functions were also determined. Moreover, the percentage of CD14+ CD16+ Mo was evaluated. We found that the entire HUS AP Mo population exhibited reduced CD14, CD64, and CD11b expression and decreased LPS-induced tumor necrosis factor production and Fcgamma-dependent cytotoxicity. HUS AP showed an increased percentage of CD14+ CD16+ Mo with higher CD16 and lower CD14 levels compared with the same subset from healthy children. Moreover, the CD14++ CD16- Mo subpopulation of HUS AP had a decreased HLA-DR expression, which correlated with severity. In conclusion, the Mo population from HUS AP patients presents phenotypic and functional alterations. The contribution to the pathogenesis and the possible scenarios that led to these changes are discussed.


Asunto(s)
Antígenos de Diferenciación/inmunología , Sangre Fetal/inmunología , Síndrome Hemolítico-Urémico/inmunología , Síndrome Hemolítico-Urémico/fisiopatología , Monocitos/inmunología , Recuento de Células , Preescolar , Citotoxicidad Inmunológica , Citometría de Flujo , Síndrome Hemolítico-Urémico/diagnóstico , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Lactante , Lipopolisacáridos/farmacología , Fenotipo , Factores de Necrosis Tumoral/inmunología
6.
Sci Rep ; 6: 24913, 2016 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-27118524

RESUMEN

Shiga toxin (Stx)-producing Escherichia coli (STEC) infections are implicated in the development of the life-threatening Hemolytic Uremic Syndrome (HUS). Despite the magnitude of the social and economic problems caused by STEC infections, no licensed vaccine or effective therapy is presently available for human use. Single chain antibodies (VHH) produced by camelids exhibit several advantages in comparison with conventional antibodies, making them promising tools for diagnosis and therapy. In the present work, the properties of a recently developed immunogen, which induces high affinity and protective antibodies against Stx type 2 (Stx2), were exploited to develop VHHs with therapeutic potential against HUS. We identified a family of VHHs against the B subunit of Stx2 (Stx2B) that neutralize Stx2 in vitro at subnanomolar concentrations. One VHH was selected and was engineered into a trivalent molecule (two copies of anti-Stx2B VHH and one anti-seroalbumin VHH). The resulting molecule presented extended in vivo half-life and high therapeutic activity, as demonstrated in three different mouse models of Stx2-toxicity: a single i.v. lethal dose of Stx2, several i.v. incremental doses of Stx2 and intragastrical STEC infection. This simple antitoxin agent should offer new therapeutic options for treating STEC infections to prevent or ameliorate HUS outcome.


Asunto(s)
Antitoxinas/aislamiento & purificación , Síndrome Hemolítico-Urémico/terapia , Inmunoterapia/métodos , Toxina Shiga II/inmunología , Anticuerpos de Cadena Única/aislamiento & purificación , Animales , Antitoxinas/uso terapéutico , Camelus , Modelos Animales de Enfermedad , Síndrome Hemolítico-Urémico/diagnóstico , Ratones , Pruebas Serológicas/métodos , Anticuerpos de Cadena Única/uso terapéutico , Terapéutica , Resultado del Tratamiento
7.
PLoS One ; 8(2): e57128, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23451160

RESUMEN

Shiga toxins (Stx) are the main virulence factors in enterohemorrhagic Escherichia coli (EHEC) infections, causing diarrhea and hemolytic uremic syndrome (HUS). The genes encoding for Shiga toxin-2 (Stx2) are located in a bacteriophage. The toxin is formed by a single A subunit and five B subunits, each of which has its own promoter sequence. We have previously reported the expression of the B subunit within the eukaryotic environment, probably driven by their own promoter. The aim of this work was to evaluate the ability of the eukaryotic machinery to recognize stx2 sequences as eukaryotic-like promoters. Vero cells were transfected with a plasmid encoding Stx2 under its own promoter. The cytotoxic effect on these cells was similar to that observed upon incubation with purified Stx2. In addition, we showed that Stx2 expression in Stx2-insensitive BHK eukaryotic cells induced drastic morphological and cytoskeletal changes. In order to directly evaluate the capacity of the wild promoter sequences of the A and B subunits to drive protein expression in mammalian cells, GFP was cloned under eukaryotic-like putative promoter sequences. GFP expression was observed in 293T cells transfected with these constructions. These results show a novel and alternative way to synthesize Stx2 that could contribute to the global understanding of EHEC infections with immediate impact on the development of treatments or vaccines against HUS.


Asunto(s)
Regiones Promotoras Genéticas , Toxina Shiga/genética , Animales , Secuencia de Bases , Diferenciación Celular , Línea Celular , Chlorocebus aethiops , Cricetinae , Homología de Secuencia de Ácido Nucleico , Células Vero
9.
J Cardiovasc Med (Hagerstown) ; 10(6): 489-91, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19365275

RESUMEN

We report a modification of the Norwood stage I procedure in a neonate with right aortic arch and complete atrio-ventricular canal in a variant of hypoplastic heart syndrome. Because of the unusual anatomy, the neo-aorta was reconstructed with a specially trimmed homograft patch and the pulmonary flow was maintained with implantation of a right ventricle to pulmonary artery shunt on the left side of the aorta. The patient had a favourable outcome and is now awaiting the stage II procedure.


Asunto(s)
Anomalías Múltiples/cirugía , Aorta Torácica/cirugía , Implantación de Prótesis Vascular , Procedimientos Quirúrgicos Cardíacos , Síndrome del Corazón Izquierdo Hipoplásico/cirugía , Aorta Torácica/anomalías , Humanos , Recién Nacido , Masculino , Venas Pulmonares/anomalías , Venas Pulmonares/cirugía , Situs Inversus/cirugía , Resultado del Tratamiento
10.
Pediatr Res ; 61(1): 123-8, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17211153

RESUMEN

Hemolytic Uremic Syndrome (HUS) is the main cause of acute renal failure in children. The high percentage of patients who develop long-term sequelae constitutes an important medical concern. The identification of parameters that correlate with the degree of renal failure may be useful to plan the best treatment soon after hospitalization. Here, we investigated the functional state of neutrophils (PMN) from HUS patients on admission, before dialysis and/or transfusion, in relation to the severity of renal impairment reached during the acute period (AP). We found that all PMN activation parameters measured in severe cases of HUS (HUS AP3) were statistically lower comparing to children with mild cases of HUS (HUS AP1). As HUS PMN phenotype and dysfunction is compatible with that of cells undergoing cell death, we also studied spontaneous apoptosis. Not only were HUS PMN not apoptotic, but HUS AP3 PMN showed an increased survival. Almost all phenotypic and functional parameters measured on PMN correlated with severity. Our results revealed a marked deactivation of PMN in severe cases of HUS, and suggest that studying the functional state of PMN could be of prognostic value.


Asunto(s)
Síndrome Hemolítico-Urémico/diagnóstico , Síndrome Hemolítico-Urémico/fisiopatología , Neutrófilos/fisiología , Preescolar , Femenino , Humanos , Lactante , Masculino , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA