Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
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.
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
3.
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
4.
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
5.
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
6.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA