Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mediators Inflamm ; 2015: 757059, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25767334

RESUMO

Although fish oil-based and olive oil-based lipid emulsions have been shown to exert anti-inflammatory functions, the immunomodulating properties of lipids are still controversial. Therefore, we investigated the anti-inflammatory effect of three different parenterally administered lipid emulsions in vivo: olive oil-based Clinoleic, fish oil-based Smoflipid, and soybean oil-based Lipofundin. We observed leukocyte recruitment in inflamed murine cremaster muscle using intravital microscopy and survival in a murine model of LPS-induced systemic inflammation and analyzed expression of leukocyte and endothelial adhesion molecules. Olive oil-based Clinoleic and fish oil-based Smoflipid profoundly inhibited leukocyte adhesion compared to Lipofundin during LPS-induced inflammation of the murine cremaster muscle. In the trauma model of cremaster muscle inflammation, Lipofundin was the only lipid emulsion that even augmented leukocyte adhesion. In contrast to Smoflipid and Lipofundin, Clinoleic effectively blocked leukocyte recruitment and increased survival during lethal endotoxemia. Flow chamber experiments and analysis of adhesion molecule expression suggest that both endothelial and leukocyte driven mechanisms might contribute to anti-inflammatory effects of Clinoleic. We conclude that the anti-inflammatory properties of Clinoleic are superior to those of Smoflipid and Lipofundin even during systemic inflammation. Thus, these results should stimulate further studies investigating parenteral lipids as an anti-inflammatory strategy in critically ill patients.


Assuntos
Leucócitos/citologia , Leucócitos/efeitos dos fármacos , Azeite de Oliva/química , Óleos de Plantas/química , Óleos de Plantas/farmacologia , Óleo de Soja/química , Óleo de Soja/farmacologia , Animais , Células Cultivadas , Óleos de Peixe/química , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Músculos/citologia , Músculos/efeitos dos fármacos
2.
Mediators Inflamm ; 2014: 283126, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25530682

RESUMO

As nuclear factor kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs) seem to be critical mediators in the inflammatory response, we studied the effects of lipopolysaccharide (LPS) and lipoteichoic acid (LTA) on (a) the activation of NF-κB and MAPKs and (b) the expression of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) with or without the specific inhibitors of these intracellular signal transduction pathways in neonatal cord and adult blood. TNF-α and IL-6 concentrations showed a sharp increase in the supernatants of cord and adult whole blood after stimulation. TNF-α concentrations were significantly higher, whereas IL-6 concentrations were tendentially lower in adult blood after stimulation. Stimulation with LPS or LTA resulted in a significantly decreased activation of p38 MAPK in neonatal compared with adult blood. Although LTA failed to induce additional ERK1/2 phosphorylation, LPS stimulation mediated the moderately increased levels of activated ERK1/2 in neonatal monocytes. The addition of the p38 MAPK inhibitor SB202190 significantly decreased IL-6 and TNF-α production upon LPS or LTA stimulation. Furthermore, the inhibition of ERK1/2 was able to reduce LPS-stimulated TNF-α production in neonatal blood. We conclude that p38 MAPK as well as ERK1/2 phosphorylation is crucially involved in LPS activation and could explain the differences in early cytokine response between neonatal and adult blood.


Assuntos
Regulação da Expressão Gênica , Interleucina-6/sangue , Lipopolissacarídeos/química , NF-kappa B/sangue , Ácidos Teicoicos/química , Fator de Necrose Tumoral alfa/sangue , Adulto , Feminino , Sangue Fetal/metabolismo , Citometria de Fluxo , Humanos , Imidazóis/química , Recém-Nascido , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Monócitos/citologia , Fosforilação , Gravidez , Piridinas/química , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
BMC Immunol ; 15: 53, 2014 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-25428166

RESUMO

BACKGROUND: Insufficient leukocyte recruitment may be one reason for the high incidence of life-threatening infections in preterm infants. Since the receptor of advanced glycation end products (RAGE) is a known leukocyte adhesion molecule and highly expressed during early development, we asked whether RAGE plays a role for leukocyte recruitment in preterm and term infants. METHODS: Leukocyte adhesion was analyzed in dynamic flow chamber experiments using isolated leukocytes of cord blood from extremely premature (<30 weeks of gestation), moderately premature (30-35 weeks of gestation) and mature neonates (>35 weeks of gestation) and compared to the results of adults. For fluorescent microscopy leukocytes were labeled with rhodamine 6G. In the respective age groups we also measured the plasma concentration of soluble RAGE (sRAGE) by ELISA and Mac-1 and LFA-1 expression on neutrophils by flow cytometry. RESULTS: The adhesive functions of fetal leukocytes significantly increase with gestational age. In all age groups, leukocyte adhesion was crucially dependent on RAGE. In particular, RAGE was equally effective to mediate leukocyte adhesion when compared to ICAM-1. The plasma levels of sRAGE were high in extremely premature infants and decreased with increasing gestational age. In contrast, expression of ß2-Integrins Mac-1 and LFA-1 which are known ligands for RAGE and ICAM-1 did not change during fetal development. CONCLUSION: We conclude that RAGE is a crucial leukocyte adhesion molecule in both preterm and term infants.


Assuntos
Lactente Extremamente Prematuro/sangue , Leucócitos/metabolismo , Receptores Imunológicos/sangue , Adulto , Adesão Celular/imunologia , Feminino , Regulação da Expressão Gênica/imunologia , Humanos , Lactente Extremamente Prematuro/imunologia , Recém-Nascido , Molécula 1 de Adesão Intercelular/sangue , Molécula 1 de Adesão Intercelular/imunologia , Leucócitos/imunologia , Leucócitos/patologia , Antígeno-1 Associado à Função Linfocitária/sangue , Antígeno-1 Associado à Função Linfocitária/imunologia , Antígeno de Macrófago 1/sangue , Antígeno de Macrófago 1/imunologia , Masculino , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/imunologia
4.
Mediators Inflamm ; 2014: 743678, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24876676

RESUMO

By binding ß 2-integrins both ICAM-1 and the receptor for advanced glycation end products (RAGE) mediate leukocyte recruitment in a stimulus-dependent manner. Using different inflammatory mouse models we investigated how RAGE and ICAM-1 are involved in anti-inflammatory functions of protein C (PC; Ceprotin, 100 U/kg). We found that, depending on the stimulus, RAGE and ICAM-1 are cooperatively involved in PC-induced inhibition of leukocyte recruitment in cremaster models of inflammation. During short-term proinflammatory stimulation (trauma, fMLP, and CXCL1), ICAM-1 is more important for mediation of anti-inflammatory effects of PC, whereas RAGE plays a major role after longer proinflammatory stimulation (TNF α ). In contrast to WT and Icam-1(-/-) mice, PC had no effect on bronchoalveolar neutrophil emigration in RAGE(-/-) mice during LPS-induced acute lung injury, suggesting that RAGE critically mediates PC effects during acute lung inflammation. In parallel, PC treatment effectively blocked leukocyte recruitment and improved survival of WT mice and Icam-1-deficient mice in LPS-induced endotoxemia, but failed to do so in RAGE-deficient mice. Exploring underlying mechanisms, we found that PC is capable of downregulating intracellular RAGE and extracellular ICAM-1 in endothelial cells. Taken together, our data show that RAGE and ICAM-1 are required for the anti-inflammatory functions of PC.


Assuntos
Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/fisiologia , Proteína C/metabolismo , Receptores Imunológicos/fisiologia , Lesão Pulmonar Aguda/patologia , Animais , Coagulação Sanguínea , Modelos Animais de Doenças , Células Endoteliais/citologia , Endotoxemia/patologia , Leucócitos/citologia , Lipopolissacarídeos/química , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Músculo Esquelético/patologia , Receptor para Produtos Finais de Glicação Avançada , Fator de Necrose Tumoral alfa/metabolismo
5.
PLoS One ; 9(2): e89422, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586767

RESUMO

AIMS: The receptor for advanced glycation endproducts, RAGE, is a multiligand receptor and NF-κB activator leading to perpetuation of inflammation. We investigated whether and how RAGE is involved in mediation of anti-inflammatory properties of protein C. METHODS AND RESULTS: We analyzed the effect of protein C on leukocyte adhesion and transmigration in WT- and RAGE-deficient mice using intravital microscopy of cremaster muscle venules during trauma- and TNFα-induced inflammation. Both, protein C (PC, Ceprotin, 100 U/kg) and activated protein C (aPC, 24 µg/kg/h) treatment significantly inhibited leukocyte adhesion in WT mice in these inflammation models. The impaired leukocyte adhesion after trauma-induced inflammation in RAGE knockout mice could not be further reduced by PC and aPC. After TNFα-stimulation, however, aPC but not PC treatment effectively blocked leukocyte adhesion in these mice. Consequently, we asked whether RAGE is involved in PC activation. Since RAGE-deficient mice and endothelial cells showed insufficient PC activation, and since thrombomodulin (TM) and endothelial protein C receptor (EPCR) are reduced on the mRNA and protein level in RAGE deficient endothelial cells, an involvement of RAGE in TM-EPCR-dependent PC activation is likely. Moreover, TNFα-induced activation of MAPK and upregulation of ICAM-1 and VCAM-1 are reduced both in response to aPC treatment and in the absence of RAGE. Thus, there seems to be interplay of the RAGE and the PC pathway in inflammation. CONCLUSION: RAGE controls anti-inflammatory properties and activation of PC, which might involve EPCR and TM.


Assuntos
Anti-Inflamatórios/metabolismo , Proteína C/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais/genética , Animais , Adesão Celular/genética , Células Endoteliais/metabolismo , Receptor de Proteína C Endotelial , Humanos , Inflamação/genética , Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Leucócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína C/genética , Receptor para Produtos Finais de Glicação Avançada , Receptores de Superfície Celular/genética , Receptores Imunológicos/genética , Trombomodulina/genética , Trombomodulina/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/genética , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
6.
Mediators Inflamm ; 2012: 739176, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23093821

RESUMO

It is well acknowledged that proinflammatory stimulation during acute hyperglycemia is able to aggravate inflammatory diseases. However, the mechanisms of proinflammatory effects of glucose are controversially discussed. We investigated leukocyte recruitment after intravenous injection of glucose in different inflammatory models using intravital microscopy. Flow chamber experiments, expression analysis, functional depletion, and knockout of key adhesion molecules gave mechanistic insight in involved pathways. We demonstrated that a single injection of glucose rapidly increased blood glucose levels in a dose-dependent manner. Notably, during tumor necrosis factor (TNF) α-induced inflammation leukocyte recruitment was not further enhanced by glucose administration, whereas glucose injection profoundly augmented leukocyte adhesion and transmigration into inflamed tissue in the trauma model, indicating that proinflammatory properties of glucose are stimulus dependent. Experiments with functional or genetic inhibition of the chemokine receptor CXCR2, intercellular adhesion molecule 1 (ICAM1), and lymphocyte function antigen 1 (LFA1) suggest that keratino-derived-chemokine CXCL1-triggered interactions of ICAM1 and LFA1 are crucially involved in the trauma model of inflammation. The lacking effect of glucose on ß(2) integrin expression and on leukocyte adhesion in dynamic flow chamber experiments argues against leukocyte-driven underlying mechanisms and favours an endothelial pathway since endothelial ICAM1 expression was significantly upregulated in response to glucose.


Assuntos
Quimiocina CXCL1/farmacologia , Glucose/farmacologia , Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Antígeno-1 Associado à Função Linfocitária/metabolismo , Animais , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Citometria de Fluxo , Imuno-Histoquímica , Molécula 1 de Adesão Intercelular/genética , Leucócitos/citologia , Antígeno-1 Associado à Função Linfocitária/genética , Camundongos , Camundongos Knockout , Ligação Proteica/efeitos dos fármacos
7.
BMC Immunol ; 12: 56, 2011 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-21970746

RESUMO

BACKGROUND: The receptor for advanced glycation endproducts, RAGE, is involved in the pathogenesis of many inflammatory conditions, which is mostly related to its strong activation of NF-κB but also due to its function as ligand for the ß2-integrin Mac-1. To further dissect the stimulus-dependent role of RAGE on leukocyte recruitment during inflammation, we investigated ß2-integrin-dependent leukocyte adhesion in RAGE-/- and Icam1-/- mice in different cremaster muscle models of inflammation using intravital microscopy. RESULTS: We demonstrate that RAGE, but not ICAM-1 substantially contributes to N-formyl-methionyl-leucyl-phenylalanine (fMLP)-induced leukocyte adhesion in TNF-α-pretreated cremaster muscle venules in a Mac-1-dependent manner. In contrast, fMLP-stimulated leukocyte adhesion in unstimulated cremaster muscle venules is independent of RAGE, but dependent on ICAM-1 and its interaction with LFA-1. Furthermore, chemokine CXCL1-stimulated leukocyte adhesion in surgically prepared cremaster muscle venules was independent of RAGE but strongly dependent on ICAM-1 and LFA-1 suggesting a differential and stimulus-dependent regulation of leukocyte adhesion during inflammation in vivo. CONCLUSION: Our results demonstrate that RAGE and ICAM-1 differentially regulate leukocyte adhesion in vivo in a stimulus-dependent manner.


Assuntos
Molécula 1 de Adesão Intercelular/metabolismo , Leucócitos/metabolismo , Antígeno-1 Associado à Função Linfocitária/metabolismo , Receptores Imunológicos/metabolismo , Vênulas/metabolismo , Animais , Adesão Celular/genética , Adesão Celular/imunologia , Células Cultivadas , Quimiocina CXCL1/metabolismo , Inflamação , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Leucócitos/imunologia , Leucócitos/patologia , Antígeno-1 Associado à Função Linfocitária/imunologia , Antígeno de Macrófago 1/imunologia , Antígeno de Macrófago 1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculos/patologia , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Ligação Proteica/genética , Ligação Proteica/imunologia , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Vênulas/patologia
8.
Am J Pathol ; 179(5): 2637-50, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21907691

RESUMO

Anti-inflammatory properties of protein C (PC) concentrate are poorly studied compared to activated protein C, although PC is suggested to be safer in clinical use. We investigated how PC interferes with the leukocyte recruitment cascade during acute inflammation and its efficacy during murine endotoxemia. We found that similar to activated protein infusion, intravenous PC application reduced leukocyte recruitment in inflamed tissues in a dose- and time-dependent manner. During both tumor necrosis factor-α induced and trauma-induced inflammation of the cremaster muscle, intravital microscopy revealed that leukocyte adhesion and transmigration, but not rolling, were profoundly inhibited by 100 U/kg PC. Moreover, PC blocked leukocyte emigration into the bronchoalveolar space during lipopolysaccharide (LPS) induced acute lung injury. PC was efficiently activated in a murine endotoxemia model, which reduced leukocyte infiltration of organs and strongly improved survival (75% versus 25% of control mice). Dependent on the inflammatory model, PC provoked a significant inhibition of leukocyte recruitment as early as 1 hour after administration. PC-induced inhibition of leukocyte recruitment during acute inflammation critically involves thrombomodulin-mediated PC activation, subsequent endothelial PC receptor and protease-activated receptor-1-dependent signaling, and down-regulation of intercellular adhesion molecule 1 leading to reduced endothelial inflammatory response. We conclude that during acute inflammation and sepsis, PC is a fast acting and effective therapeutic approach to block leukocyte recruitment and improve survival.


Assuntos
Lesão Pulmonar Aguda/imunologia , Anti-Inflamatórios/farmacologia , Endotoxemia/imunologia , Leucócitos/efeitos dos fármacos , Pneumonia/imunologia , Proteína C/farmacologia , Animais , Antibacterianos/farmacologia , Antígenos CD/metabolismo , Adesão Celular/imunologia , Movimento Celular/imunologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/imunologia , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Receptor de Proteína C Endotelial , Endotoxemia/tratamento farmacológico , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/imunologia , Molécula 1 de Adesão Intercelular/fisiologia , Lipopolissacarídeos/toxicidade , Antígeno-1 Associado à Função Linfocitária/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/imunologia , Músculo Liso Vascular/lesões , Miosite/imunologia , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Análise de Sobrevida , Trombomodulina/metabolismo , Fator de Necrose Tumoral alfa/toxicidade
9.
Blood ; 116(5): 841-9, 2010 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-20407037

RESUMO

The receptor for advanced glycation end products (RAGE) contributes to the inflammatory response in many acute and chronic diseases. In this context, RAGE has been identified as a ligand for the beta(2)-integrin Mac-1 under static in vitro conditions. Because intercellular adhesion molecule (ICAM)-1 also binds beta(2)-integrins, we studied RAGE(-/-), Icam1(-/-), and RAGE(-/-) Icam1(-/-) mice to define the relative contribution of each ligand for leukocyte adhesion in vivo. We show that trauma-induced leukocyte adhesion in cremaster muscle venules is strongly dependent on RAGE and ICAM-1 acting together in an overlapping fashion. Additional in vivo experiments in chimeric mice lacking endothelium-expressed RAGE and ICAM-1 located the adhesion defect to the endothelial compartment. Using microflow chambers coated with P-selectin, CXCL1, and soluble RAGE (sRAGE) demonstrated that sRAGE supports leukocyte adhesion under flow conditions in a Mac-1- but not LFA-1-dependent fashion. A static adhesion assay revealed that wild-type and RAGE(-/-) neutrophil adhesion and spreading were similar on immobilized sRAGE or fibrinogen. These observations indicate a crucial role of endothelium-expressed RAGE as Mac-1 ligand and uncover RAGE and ICAM-1 as a new set of functionally linked adhesion molecules, which closely cooperate in mediating leukocyte adhesion during the acute trauma-induced inflammatory response in vivo.


Assuntos
Quimiotaxia de Leucócito/fisiologia , Endotélio Vascular/metabolismo , Molécula 1 de Adesão Intercelular/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Músculo Esquelético/irrigação sanguínea , Vasculite/imunologia , Doença Aguda , Animais , Adesão Celular , Forma Celular , Quimiotaxia de Leucócito/efeitos dos fármacos , Humanos , Leucotrieno B4/farmacologia , Ligantes , Antígeno de Macrófago 1/metabolismo , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/deficiência , Músculo Esquelético/lesões , Músculo Esquelético/patologia , Neutrófilos/patologia , Quimera por Radiação , Proteínas Recombinantes de Fusão/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Vasculite/etiologia , Vênulas/patologia
10.
Am J Physiol Lung Cell Mol Physiol ; 298(3): L462-71, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20061440

RESUMO

Acute lung injury (ALI) still poses a major challenge in critical care medicine. Neutrophils, platelets, and chemokines are all considered key components in the development of ALI. The Duffy antigen receptor for chemokines (DARC) is thought to be involved in scavenging, transendothelial transport, and presentation of neutrophil-specific chemokines. DARC is expressed on endothelial cells and erythrocytes but not on leukocytes. Here, we show that DARC is crucial for chemokine-mediated leukocyte recruitment in vivo. However, we also demonstrate that changes in chemokine and chemokine receptor homeostasis, associated with Darc gene deficiency, exert strong anti-inflammatory effects. Neutrophils from Darc gene-deficient (Darc(-/-)) mice display a more prolonged downregulation of CXCR2 during severe inflammation than neutrophils from wild-type mice. In a CXCR2-dependent model of acid-induced ALI, Darc gene deficiency prevents ALI. Darc(-/-) mice demonstrate fully preserved oxygenation, only a small increase in vascular permeability, and a complete lack of pulmonary neutrophil recruitment. Further analysis reveals that only neutrophils but neither endothelial cells nor erythrocytes from Darc(-/-) mice confer protection from ALI. The protection appears to be due to abolished pulmonary recruitment of neutrophils from Darc(-/-) mice. The generation of neutrophil-platelet aggregates, a key mechanism in both pulmonary neutrophil recruitment and thrombus formation, is also affected by altered CXCR2 homeostasis in Darc(-/-) mice. CXCR2 blockade enhances the formation of platelet-neutrophil aggregates and thereby corrects a formerly unknown bleeding defect in Darc(-/-) mice. In summary, our study suggests that chemokine/chemokine receptor homeostasis plays a previously unrecognized and crucial role in severe ALI.


Assuntos
Lesão Pulmonar Aguda , Quimiocinas , Sistema do Grupo Sanguíneo Duffy , Homeostase , Receptores de Superfície Celular , Animais , Camundongos , Lesão Pulmonar Aguda/sangue , Lesão Pulmonar Aguda/complicações , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/prevenção & controle , Tempo de Sangramento , Contagem de Células Sanguíneas , Agregação Celular , Quimiocina CXCL1/sangue , Quimiocinas/metabolismo , Quimiotaxia de Leucócito , Regulação para Baixo , Sistema do Grupo Sanguíneo Duffy/genética , Sistema do Grupo Sanguíneo Duffy/metabolismo , Células Endoteliais/citologia , Eritrócitos/citologia , Hemorragia/complicações , Hemorragia/patologia , Ácido Clorídrico , Inflamação/metabolismo , Inflamação/patologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Agregação Plaquetária , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Interleucina-8B/metabolismo
11.
Crit Care Med ; 35(9): 2156-63, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17855830

RESUMO

OBJECTIVE: Acute renal failure remains a major challenge in critical care medicine. Both neutrophils and chemokines have been proposed as key components in the development of acute renal failure. Although the Duffy antigen receptor for chemokines (DARC) is present in several tissues and a highly specific ligand for various chemokines, its exact role in vivo remains unclear. DESIGN: Prospective, controlled experimental study. SETTING: University-based research laboratory. SUBJECTS: C57BL/6 wild-type and DARC gene-deficient mice (DARC-/-). INTERVENTIONS: To unravel the functional relevance of DARC in vivo, we compared wild-type and DARC-/- using neutrophil-dependent models of acute renal failure, induced by either local (renal ischemia-reperfusion) or systemic (endotoxemia, lipopolysaccharide) injury. MEASUREMENTS AND MAIN RESULTS: Plasma creatinine and blood urea nitrogen concentrations served as indicators of renal function or dysfunction. Enzyme-linked immunosorbent assays were used to measure tissue and plasma chemokine concentrations. We also performed immunostaining to localize chemokine expression and flow cytometry to evaluate neutrophil recruitment into the kidney. Following renal injury, wild-type mice developed moderate renal ischemia-reperfusion(lipopolysaccharide, 300% increase in plasma creatinine concentrations) to severe acute renal failure (renal ischemia-reperfusion, 40% mortality) as well as extensive renal neutrophil recruitment. DARC-/- mice exhibited no renal dysfunction (renal ischemia-reperfusion) or only very mild renal dysfunction (lipopolysaccharide, 20% increase in serum creatinine concentrations). DARC-/- mice showed no postischemic neutrophil infiltration. Although DARC-/- and wild-type mice exhibited similar global renal neutrophil-recruitment during endotoxemia, DARC-/- mice showed significantly impaired neutrophil extravasation. Total renal concentrations of the chemokine macrophage inflammatory protein 2, which has been shown to bind to DARC and to be crucial in postischemic acute renal failure, were either identical (lipopolysaccharide) or only moderately different (renal ischemia-reperfusion) between wild-type and DARC-/- mice. Immunostaining revealed an absence of macrophage inflammatory protein-2 in renal endothelial cells of DARC-/- mice. CONCLUSIONS: We suggest that DARC predominantly exerts its effects by controlling spatial chemokine distribution, which in turn regulates neutrophil recruitment and subsequent acute renal failure.


Assuntos
Injúria Renal Aguda , Sistema do Grupo Sanguíneo Duffy , Receptores de Superfície Celular , Animais , Camundongos , Injúria Renal Aguda/fisiopatologia , Nitrogênio da Ureia Sanguínea , Quimiocinas/análise , Creatinina/sangue , Sistema do Grupo Sanguíneo Duffy/genética , Sistema do Grupo Sanguíneo Duffy/fisiologia , Ensaio de Imunoadsorção Enzimática , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos/fisiologia , Estudos Prospectivos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...