Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Pediatr Nephrol ; 33(11): 2057-2071, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29372302

RESUMO

Hemolytic uremic syndrome (HUS) is defined as a triad of noninmune microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury. The most frequent presentation is secondary to Shiga toxin (Stx)-producing Escherichia coli (STEC) infections, which is termed postdiarrheal, epidemiologic or Stx-HUS, considering that Stx is the necessary etiological factor. After ingestion, STEC colonize the intestine and produce Stx, which translocates across the intestinal epithelium. Once Stx enters the bloodstream, it interacts with renal endothelial and epithelial cells, and leukocytes. This review summarizes the current evidence about the involvement of inflammatory components as central pathogenic factors that could determine outcome of STEC infections. Intestinal inflammation may favor epithelial leakage and subsequent passage of Stx to the systemic circulation. Vascular damage triggered by Stx promotes not only release of thrombin and increased fibrin concentration but also production of cytokines and chemokines by endothelial cells. Recent evidence from animal models and patients strongly indicate that several immune cells types may participate in HUS physiopathology: neutrophils, through release of proteases and reactive oxygen species (ROS); monocytes/macrophages through secretion of cytokines and chemokines. In addition, high levels of Bb factor and soluble C5b-9 (sC5b-9) in plasma as well as complement factors adhered to platelet-leukocyte complexes, microparticles and microvesicles, suggest activation of the alternative pathway of complement. Thus, acute immune response secondary to STEC infection, the Stx stimulatory effect on different immune cells, and inflammatory stimulus secondary to endothelial damage all together converge to define a strong inflammatory status that worsens Stx toxicity and disease.


Assuntos
Infecções por Escherichia coli/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Microvasos/patologia , Escherichia coli Shiga Toxigênica/imunologia , Animais , Via Alternativa do Complemento/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/patologia , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Células Epiteliais/imunologia , Células Epiteliais/patologia , Infecções por Escherichia coli/sangue , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Síndrome Hemolítico-Urêmica/sangue , Síndrome Hemolítico-Urêmica/microbiologia , Síndrome Hemolítico-Urêmica/patologia , Humanos , Mucosa Intestinal/microbiologia , Rim/irrigação sanguínea , Rim/imunologia , Rim/patologia , Microvasos/citologia , Microvasos/imunologia , Escherichia coli Shiga Toxigênica/isolamento & purificação
2.
Toxins (Basel) ; 9(11)2017 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-29068360

RESUMO

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.


Assuntos
Ligante de CD40/sangue , Células Endoteliais/efeitos dos fármacos , Síndrome Hemolítico-Urêmica/sangue , Toxina Shiga/toxicidade , Células Cultivadas , Criança , Pré-Escolar , Células Endoteliais/patologia , Feminino , Síndrome Hemolítico-Urêmica/induzido quimicamente , Humanos , Lactente , Rim/metabolismo , Rim/patologia , Masculino , Microvasos , Monócitos/metabolismo , Estresse Oxidativo , Ativação Plaquetária/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
3.
J Innate Immun ; 8(4): 400-11, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27230920

RESUMO

Hemolytic uremic syndrome (HUS), a vascular disease characterized by hemolytic anemia, thrombocytopenia, and acute renal failure, is caused by enterohemorrhagic Shiga toxin (Stx)-producing bacteria, which mainly affect children. Besides Stx, the inflammatory response mediated by neutrophils (PMN) is essential to HUS evolution. PMN can release neutrophil extracellular traps (NET) composed of DNA, histones, and other proteins. Since NET are involved in infectious and inflammatory diseases, the aim of this work was to investigate the contribution of NET to HUS. Plasma from HUS patients contained increased levels of circulating free-DNA and nucleosomes in comparison to plasma from healthy children. Neutrophils from HUS patients exhibited a greater capacity to undergo spontaneous NETosis. NET activated human glomerular endothelial cells, stimulating secretion of the proinflammatory cytokines IL-6 and IL-8. Stx induced PMN activation as judged by its ability to trigger reactive oxygen species production, increase CD11b and CD66b expression, and induce NETosis in PMN from healthy donors. During HUS, NET can contribute to the inflammatory response and thrombosis in the microvasculature and thus to renal failure. Intervention strategies to inhibit inflammatory mechanisms mediated by PMN, such as NETosis, could have a potential therapeutic impact towards amelioration of the severity of HUS.


Assuntos
Infecções Bacterianas/imunologia , Células Endoteliais/imunologia , Armadilhas Extracelulares/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Rim/patologia , Neutrófilos/imunologia , Toxina Shiga/imunologia , Injúria Renal Aguda , Anemia Hemolítica , Apoptose , Células Cultivadas , Criança , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Ativação de Neutrófilo , Neutrófilos/microbiologia , Espécies Reativas de Oxigênio/metabolismo , Trombocitopenia
5.
J Clin Immunol ; 32(3): 622-31, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22228570

RESUMO

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.


Assuntos
Síndrome Hemolítico-Urêmica/imunologia , Interleucina-10/imunologia , Monócitos/imunologia , Fator de Necrose Tumoral alfa/imunologia , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Interleucina-10/sangue , Lipopolissacarídeos/imunologia , Masculino , Fator de Necrose Tumoral alfa/sangue
6.
Medicina (B.Aires) ; 71(4): 383-389, July-Aug. 2011. ilus
Artigo em Espanhol | LILACS | ID: lil-633884

RESUMO

La forma típica o post-diarreica del síndrome urémico hemolítico (SUH) es la complicación más grave de las infecciones por cepas de Escherichia coli productoras de toxina Shiga (STEC). En la Argentina el SUH es un problema crítico de salud pública, ya que representa la principal causa de falla renal aguda en la infancia, la segunda causa de falla renal crónica, y aporta el 20% de los casos de transplante renal durante la infancia y la adolescencia. A pesar de los avances en el conocimiento de su patogénesis, el único tratamiento actual de los pacientes con SUH es de sostén, y no existen terapias específicas ni preventivas. En la presente revisión expondremos los conocimientos básicos de los mecanismos patogénicos y discutiremos los enfoques terapéuticos tradicionales e innovadores, con especial foco en la situación nacional y los aportes hechos por grupos de la Argentina.


The typical form of hemolytic uremic syndrome (HUS) is the major complication of Shiga toxin-producing Escherichia coli (STEC) infections. HUS is a critical health problem in Argentina since it is the main cause of acute renal failure in children and the second cause of chronic renal failure, giving account for 20% of renal transplants in children and adolescents in our country. In spite of the extensive research in the field, the mainstay of treatment for patients with HUS is supportive therapy, and there are no specific therapies preventing or ameliorating the disease course. In this review, we present the current knowledge about pathogenic mechanisms and discuss traditional and innovative therapeutic approaches, with special focus in national status and contributions made by Argentinean groups.


Assuntos
Humanos , Síndrome Hemolítico-Urêmica , Escherichia coli Shiga Toxigênica , Argentina/epidemiologia , Síndrome Hemolítico-Urêmica/epidemiologia , Síndrome Hemolítico-Urêmica/microbiologia , Síndrome Hemolítico-Urêmica/terapia , Escherichia coli Shiga Toxigênica/patogenicidade
8.
Arch. argent. pediatr ; 106(2): 102-109, abr.2008. graf, tab
Artigo em Espanhol | LILACS | ID: lil-482394

RESUMO

Objetivo. Comparar la composición corporal estimadapor dos métodos antropométricos simples ypor impedanciometría (BIA, por sus siglas en inglés)y absorciometría de doble haz de rayos X(DXA, por sus siglas en inglés) y estudiar las correlacionesexistentes entre ellos, en una poblaciónpreescolar de Argentina.Población, material y métodos. Se estudió transversalmenteuna población clínicamente sana, deedad comprendida entre 4 y 6 años, de 230 niños(118 varones y 112 niñas), que concurrían a JardinesIntegrales de La Matanza, Argentina. Se determinóel índice de masa corporal (IMC), la circunferenciade cintura (Cci) y el contenido de grasa corporal,expresado como porcentaje, por BIA (Maltron BF-900) y por DXA (densitómetro Lunar DPX-L, softwarepediátrico).Resultados. En IMC y Cci no se observaron diferenciasestadísticamente significativas entre ambossexos, pero sí en el porcentaje de grasa corporal porBIA o DXA. La correlación entre BIA y ambos métodosantropométricos fue moderada (r de Pearson=0,43-0,53), al igual que entre DXA y Cci (r= 0,66),mientras que las demás correlaciones fueron fuertes(r= 0,71-0,83). Las medidas obtenidas por BIA yDXA no concordaron (prueba de Bland Altman); sinembargo, las diferencias se distribuyeron en formahomogénea a lo largo del eje horizontal e independientementede las medias. BIA reprodujo valoresmás bajos de porcentaje de grasa corporal que DXA(p <0,0001).Conclusiones. Existe buena correlación entre losmétodos antropométricos sencillos y la bioimpedanciay DXA. Sin embargo, los resultados no sonintercambiables, incluso entre BIA y DXA


Objective. To examine the interrelationships of body composition variables derived from simple anthropometry, bioelectrical impedance (BIA) and dualenergy X-ray absorptiometry (DXA). Population, material and methods. A total of 230 healthy preschool children (118 males and 112 females) age 4-6 years, attending a day care center, were examined to determine body mass index (BMI) and waist circumference (WC). Percentage of body fat content (%BF) was predicted by BIA (Maltron BF- 900) and DXA (Lunar DPX-L, pediatric software). Results. BMI and WC did not show significant differences among sex, while %BF by BIA or DXA did. BIA measures were lower than DXA´s (p <0.0001). Correlation between BIA vs. anthropometric methods and WC vs. DXA were moderate (Pearson r= 0.43 to 0.53), whereas the other correlations were strong (r= 0.71 to 0.83). However Bland Altman comparison showed wide limits of agreementbetween BIA and DXA; BIA significantly underestimated %BF as determined by DXA (p <0.0001). Conclusion. The methods used to estimate body composition in healthy preschool children highly correlated. However, the Bland Altman procedure suggests that BIA and DXA should not be used interchangeably.


Assuntos
Pré-Escolar , Criança , Absorciometria de Fóton , Antropometria , Composição Corporal , Índice de Massa Corporal , Peso-Idade , Peso-Estatura , Estudo Comparativo , Estudos Transversais , Epidemiologia Descritiva , Estudos Observacionais como Assunto , Interpretação Estatística de Dados
11.
Blood ; 109(6): 2438-45, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17132725

RESUMO

Thrombotic microangiopathy and acute renal failure are cardinal features of postdiarrheal hemolytic uremic syndrome (HUS). These conditions are related to endothelial and epithelial cell damage induced by Shiga toxin (Stx) through the interaction with its globotriaosyl ceramide receptor. However, inflammatory processes contribute to the pathogenesis of HUS by sensitizing cells to Stx fractalkine (FKN), a CX(3)C transmembrane chemokine expressed on epithelial and endothelial cells upon activation, is involved in the selective migration and adhesion of specific leukocyte subsets to tissues. Here, we demonstrated a selective depletion of circulating mononuclear leukocytes expressing the receptor for FKN (CX(3)CR1) in patients with HUS. We found a unique phenotype in children with HUS distinct from that seen in healthy, uremic, or infected controls, in which monocytes lost CX(3)CR1, down-modulated CD62L, and increased CD16. In addition, the CD56(dim) natural killer (NK) subpopulation was decreased, leading to an altered peripheral CD56(dim)/CD56(bright) ratio from 10.0 to 4.5. It is noteworthy that a negative correlation existed between the percentage of circulating CX(3)CR1(+) leukocytes and the severity of renal failure. Finally, CX(3)CR1(+) leukocytes were observed in renal biopsies from patients with HUS. We suggest that the interaction of CX(3)CR1(+) cells with FKN present on activated endothelial cells may contribute to renal injury in HUS.


Assuntos
Quimiocinas CX3C/metabolismo , Síndrome Hemolítico-Urêmica/etiologia , Síndrome Hemolítico-Urêmica/metabolismo , Proteínas de Membrana/metabolismo , Transdução de Sinais , Biópsia , Receptor 1 de Quimiocina CX3C , Quimiocina CX3CL1 , Pré-Escolar , Feminino , Síndrome Hemolítico-Urêmica/patologia , Síndrome Hemolítico-Urêmica/cirurgia , Humanos , Imuno-Histoquímica , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Selectina L/metabolismo , Contagem de Leucócitos , Masculino , Monócitos/metabolismo , Receptores de Quimiocinas/metabolismo
14.
Arch. latinoam. nefrol. pediátr ; 6(3): 122-122, 2006.
Artigo em Espanhol | LILACS | ID: lil-480076
15.
Medicina (B.Aires) ; 66(supl.2): 6-10, 2006. tab
Artigo em Espanhol | LILACS | ID: lil-480131

RESUMO

Clinical manifestation are described in children with epidemic HUS. The intestinal involvement in the prodromic period, is outlined and the most common disturbances such acute renal failure, thrombocytopenia, hemolytic anemia, leucocitosis hypertension, neurological, pancreatic and cardiac manifestations are described. We discuss the acid-base and electrolyte disturbances, metabolic acidosis, hyponatremia, hyperkalemia. The etiopathogenic treatment and the control of renal sequelae are also discussed.


Assuntos
Humanos , Masculino , Feminino , Lactente , Pré-Escolar , Criança , Síndrome Hemolítico-Urêmica/complicações , Síndrome Hemolítico-Urêmica/terapia , Doença Aguda , Injúria Renal Aguda , Diarreia/complicações , Diarreia/fisiopatologia , /imunologia , Vacinas contra Escherichia coli/uso terapêutico , Síndrome Hemolítico-Urêmica/fisiopatologia , Hipertensão/etiologia , Transplante de Rim , Prognóstico , Trombocitopenia/etiologia , Trombocitopenia/fisiopatologia
17.
J Leukoc Biol ; 78(4): 853-61, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16046554

RESUMO

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.


Assuntos
Antígenos de Diferenciação/imunologia , Sangue Fetal/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/fisiopatologia , Monócitos/imunologia , Contagem de Células , Pré-Escolar , Citotoxicidade Imunológica , Citometria de Fluxo , Síndrome Hemolítico-Urêmica/diagnóstico , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Lactente , Lipopolissacarídeos/farmacologia , Fenótipo , Fatores de Necrose Tumoral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA