Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Crit Care Med ; 40(2): 379-87, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22020238

RESUMO

OBJECTIVES: Matrix metalloproteinase-8 messenger RNA expression was previously found to be increased in whole blood of children with septic shock. The impact of this finding on the severity and inflammatory response to sepsis is unknown. Here, we investigate the relationship between matrix metalloproteinase-8 and disease severity in children with septic shock. We further corroborate the role of matrix metalloproteinase-8 in sepsis in a murine model. DESIGN: Retrospective observational clinical study and randomized controlled laboratory experiments. SETTING: Pediatric intensive care units and an animal research facility at an academic children's hospital. PATIENTS AND SUBJECTS: Patients age ≤10 yrs admitted to the intensive care unit with a diagnosis of septic shock. For laboratory studies, we utilized male mice deficient for matrix metalloproteinase-8 and male wild-type C57BL/6J mice. INTERVENTIONS: Blood from children with septic shock was analyzed for matrix metalloproteinase-8 messenger RNA expression and matrix metalloproteinase-8 activity, and correlated with disease severity based on mortality and degree of organ failure. A murine model of sepsis was used to explore the effect of genetic and pharmacologic inhibition of matrix metalloproteinase-8 on the inflammatory response to sepsis. Finally, activation of nuclear factor-κB was assessed both in vitro and in vivo. MEASUREMENTS AND MAIN RESULTS: Increased matrix metalloproteinase-8 mRNA expression and activity in septic shock correlates with decreased survival and increased organ failure in pediatric patients. Genetic and pharmacologic inhibition of matrix metalloproteinase-8 leads to improved survival and a blunted inflammatory profile in a murine model of sepsis. We also identify matrix metalloproteinase-8 as a direct in vitro activator of the proinflammatory transcription factor, nuclear factor-κB. CONCLUSIONS: Matrix metalloproteinase-8 is a novel modulator of inflammation during sepsis and a potential therapeutic target.


Assuntos
Mediadores da Inflamação/sangue , Metaloproteinase 8 da Matriz/sangue , Insuficiência de Múltiplos Órgãos/sangue , Choque Séptico/sangue , Choque Séptico/mortalidade , Animais , Células Cultivadas , Criança , Pré-Escolar , Estudos de Coortes , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Unidades de Terapia Intensiva Pediátrica , Leucócitos Mononucleares/metabolismo , Macrófagos Peritoneais/metabolismo , Masculino , Metaloproteinase 8 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Insuficiência de Múltiplos Órgãos/mortalidade , Prognóstico , Estudos Retrospectivos , Sepse/sangue , Sepse/tratamento farmacológico , Sepse/fisiopatologia , Índice de Gravidade de Doença , Choque Séptico/fisiopatologia , Análise de Sobrevida , Resultado do Tratamento
2.
Am J Physiol Lung Cell Mol Physiol ; 300(5): L730-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21398498

RESUMO

C-peptide is a 31-amino acid peptide cleaved from proinsulin during insulin synthesis. Initially thought to be inert, C-peptide may modulate the inflammatory response in the setting of endotoxemia and ischemia reperfusion. However, the spectrum of its biological effects is unclear. We hypothesized that exogenous administration of C-peptide would modulate pro- and anti-inflammatory signaling pathways and thereby attenuate lung inflammation in an in vivo model of hemorrhagic shock. Hemorrhagic shock was induced in male Wistar rats (aged 3-4 mo) by withdrawing blood to a mean arterial pressure of 50 mmHg. At 3 h after hemorrhage, rats were rapidly resuscitated by returning their shed blood. At the time of resuscitation and every hour thereafter, animals received C-peptide (280 nmol/kg) or vehicle parenterally. Animals were euthanized at 1 and 3 h after resuscitation. C-peptide administration at resuscitation following hemorrhagic shock ameliorated hypotension and blunted the systemic inflammatory response by reducing plasma levels of IL-1, IL-6, macrophage inflammatory protein-1α, and cytokine-induced neutrophil chemoattractant-1. This was associated with a reduction in lung neutrophil infiltration and plasma levels of receptor for advanced glycation end products. Mechanistically, C-peptide treatment was associated with reduced expression of proinflammatory transcription factors activator protein-1 and NF-κB and activation of the anti-inflammatory transcription factor peroxisome proliferator-activated receptor-γ. Our data suggest that C-peptide ameliorates the inflammatory response and lung inflammation following hemorrhagic shock. These effects may be modulated by altering the balance between pro- and anti-inflammatory signaling in the lung.


Assuntos
Peptídeo C/farmacologia , Pneumonia/prevenção & controle , Choque Hemorrágico/complicações , Animais , Citocinas/sangue , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Infiltração de Neutrófilos/efeitos dos fármacos , PPAR gama/metabolismo , Pneumonia/patologia , Ratos , Ratos Wistar , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/sangue , Ressuscitação , Choque Hemorrágico/tratamento farmacológico , Fator de Transcrição AP-1/metabolismo , Fator de Necrose Tumoral alfa/sangue
3.
Am J Pathol ; 177(4): 1834-47, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20709805

RESUMO

The nuclear peroxisome proliferator-activated receptor δ (PPARδ) is an important regulator of lipid metabolism. In contrast to its known effects on energy homeostasis, its biological role on inflammation is not well understood. We investigated the role of PPARδ in the modulation of the nuclear factor-κB (NF-κB)-driven inflammatory response to polymicrobial sepsis in vivo and in macrophages in vitro. We demonstrated that administration of GW0742, a specific PPARδ ligand, provided beneficial effects to rats subjected to cecal ligation and puncture, as shown by reduced systemic release of pro-inflammatory cytokines and neutrophil infiltration in lung, liver, and cecum, when compared with vehicle treatment. Molecular analysis revealed that treatment with GW0742 reduced NF-κB binding to DNA in lung and liver. In parallel experiments, heterozygous PPARδ-deficient mice suffered exaggerated lethality when subjected to cecal ligation and puncture and exhibited severe lung injury and higher levels of circulating tumor necrosis factor-α (TNFα) and keratinocyte-derived chemokine than wild-type mice. Furthermore, in lipopolysaccharide-stimulated J774.A1 macrophages, GW0742 reduced TNFα production by inhibiting NF-κB activation. RNA silencing of PPARδ abrogated the inhibitory effects of GW0742 on TNFα production. Chromatin immunoprecipitation assays revealed that PPARδ displaced the NF-κB p65 subunit from the κB elements of the TNFα promoter, while recruiting the co-repressor BCL6. These data suggest that PPARδ is a crucial anti-inflammatory regulator, providing a basis for novel sepsis therapies.


Assuntos
Bacteriemia/prevenção & controle , Inflamação/prevenção & controle , NF-kappa B/metabolismo , PPAR delta/fisiologia , Sepse/metabolismo , Sepse/microbiologia , Animais , Bacteriemia/etiologia , Bacteriemia/metabolismo , Western Blotting , Ceco/imunologia , Ceco/metabolismo , Ceco/microbiologia , Núcleo Celular/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Ensaio de Desvio de Mobilidade Eletroforética , Ensaio de Imunoadsorção Enzimática , Hipotensão , Técnicas Imunoenzimáticas , Inflamação/etiologia , Inflamação/metabolismo , Luciferases/metabolismo , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Knockout , NF-kappa B/genética , PPAR delta/agonistas , PPAR delta/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Sepse/imunologia , Transdução de Sinais , Taxa de Sobrevida , Tiazóis/farmacologia
4.
Am J Physiol Gastrointest Liver Physiol ; 298(1): G133-41, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19926821

RESUMO

A clinical observation in pediatric and adult intensive care units is that the incidence of multiple organ failure in pediatric trauma victims is lower than in adult patients. However, the molecular mechanisms are not yet defined. Recent experimental studies have shown that the nuclear peroxisome proliferator-activated receptor-gamma (PPARgamma) modulates the inflammatory process. In this study, we hypothesized that severity of liver injury may be age dependent and PPARgamma activation may provide beneficial effects. Hemorrhagic shock was induced in anesthetized young (3-5 mo old) and mature male Wistar rats (11-13 mo old) by withdrawing blood to a mean arterial blood pressure of 50 mmHg. After 3 h, rats were rapidly resuscitated with shed blood. Animals were euthanized 3 h after resuscitation. In mature rats, liver injury appeared more pronounced compared with young rats and was characterized by marked hepatocyte apoptosis, extravasation of erythrocytes, and accumulation of neutrophils. The ratio between the antiapoptotic protein Bcl-2 and the proapoptotic protein BAX was lower, whereas activity of caspase-3, the executioner of apoptosis, was higher in liver of mature rats compared with young rats. Plasma alanine aminotransferase levels were not different between the two age groups. This heightened liver apoptosis was associated with a significant downregulation of PPARgamma DNA binding in mature rats compared with young rats. Treatment with the PPARgamma ligand ciglitazone significantly reduced liver apoptosis in mature rats. Our data suggest that liver injury after severe hemorrhage is age dependent and PPARgamma activation is a novel hepatoprotective mechanism.


Assuntos
Apoptose/fisiologia , Hepatopatias/metabolismo , Hepatopatias/patologia , PPAR gama/metabolismo , Choque Hemorrágico/metabolismo , Choque Hemorrágico/patologia , Fatores Etários , Alanina Transaminase/sangue , Animais , Pressão Sanguínea , Caspase 3/metabolismo , Regulação para Baixo/fisiologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Hipoglicemiantes/farmacologia , Hepatopatias/tratamento farmacológico , Masculino , Neutrófilos/patologia , PPAR gama/genética , Peroxidase/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Wistar , Ressuscitação , Índice de Gravidade de Doença , Tiazolidinedionas/farmacologia , Proteína X Associada a bcl-2/metabolismo
5.
Mol Med ; 16(11-12): 491-7, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20809049

RESUMO

Peroxisome proliferator-activated receptor (PPAR)-γ is a ligand-activated transcription factor and regulates inflammation. Posttranslational modifications regulate the function of PPARγ, potentially affecting inflammation. PPARγ contains a mitogen-activated protein kinase (MAPK) site, and phosphorylation by extracellular signal-regulated kinase (ERK)-1/2 leads to inhibition of PPARγ. This study investigated the kinetics of PPARγ expression and activation in parenchymal and immune cells in sepsis using the MAPK/ERK kinase (MEK)-1 inhibitor, an upstream kinase of ERK1/2. Adult male Sprague Dawley rats were subjected to polymicrobial sepsis by cecal ligation and puncture. Rats received intraperitoneal injection of vehicle or the MEK1 inhibitor PD98059 (5 mg/kg) 30 min before cecal ligation and puncture. Rats were euthanized at 0, 1, 3, 6 and 18 h after cecal ligation and puncture. Control animals used were animals at time 0 h. Lung, plasma and peripheral blood mononuclear cells (PBMCs) were collected for biochemical assays. In vehicle-treated rats, polymicrobial sepsis resulted in significant lung injury. In the lung and PBMCs, nuclear levels of PPARγ were decreased and associated with an increase in phosphorylated PPARγ and phosphorylated ERK1/2 levels. Treatment with the MEK1 inhibitor increased the antiinflammatory plasma adipokine adiponectin, restored PPARγ expression in PBMCs and lung, and decreased lung injury. The inflammatory effects of sepsis cause changes in PPARγ expression and activation, in part, because of phosphorylation of PPARγ by ERK1/2. This phosphorylation can be reversed by ERK1/2 inhibition, thereby improving lung injury.


Assuntos
Regulação para Baixo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , PPAR gama/metabolismo , Sepse/microbiologia , Adiponectina/sangue , Animais , Anti-Inflamatórios/farmacocinética , Ligantes , Pulmão/fisiopatologia , Masculino , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Sepse/metabolismo , Fatores de Transcrição/metabolismo
6.
Crit Care Med ; 36(10): 2849-57, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18828195

RESUMO

OBJECTIVE: Peroxisome proliferator-activated receptor-gamma is a ligand-activated transcription factor. Ciglitazone, a peroxisome proliferator-activated receptor-gamma ligand, has been shown to provide beneficial effects in experimental models of sepsis and ischemia/reperfusion injury. We investigated the effects of ciglitazone on lung inflammation after severe hemorrhage. DESIGN: Prospective, laboratory study, rodent model of hemorrhagic shock. SETTING: University hospital laboratory. SUBJECTS: Male rats. INTERVENTIONS: Hemorrhagic shock was induced by withdrawing blood to a mean arterial pressure of 50 mm Hg. At 3 hrs after hemorrhage, rats were rapidly resuscitated by returning their shed blood. At the time of resuscitation and every hour thereafter, animals received ciglitazone (10 mg/kg) or vehicle intraperitoneally. Heart rate and mean arterial pressure were measured throughout the experiment. Plasma and lung tissue were collected for analysis up to 3 hrs after resuscitation. MEASUREMENTS AND MAIN RESULTS: Ciglitazone treatment ameliorated mean arterial pressure, reduced lung injury, significantly blunted lung neutrophil infiltration, and lowered plasma interleukin-6, interleukin-10, and monocyte chemoattractant protein-1 levels. In a time course analysis, vehicle-treated rats had a significant increase in nuclear factor-kappaB DNA binding, which was preceded by increased inhibitor kappaB protein kinase activity and inhibitor kappaB alpha degradation in the lung. Treatment with ciglitazone significantly reduced inhibitor kappaB protein kinase activity and inhibitor kappaB alpha degradation and completely inhibited nuclear factor-kappaB DNA binding. This reduction of inhibitor kappaB protein kinase activity afforded by ciglitazone appeared to be a consequence of a physical interaction between peroxisome proliferator-activated receptor-gamma and increased inhibitor kappaB protein kinase. CONCLUSION: Ciglitazone ameliorates the inflammatory response and may reduce lung injury after hemorrhagic shock. These protective effects appear to be mediated through inhibition of the inhibitor kappaB protein kinase/nuclear factor-kappaB pathway.


Assuntos
Mediadores da Inflamação/sangue , NF-kappa B/metabolismo , PPAR gama/metabolismo , Pneumonia/prevenção & controle , Choque Hemorrágico/tratamento farmacológico , Tiazolidinedionas/farmacologia , Animais , Glicemia/análise , Quimiocinas/sangue , Citocinas/sangue , Modelos Animais de Doenças , Imuno-Histoquímica , Infusões Parenterais , Masculino , Redes e Vias Metabólicas/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , PPAR gama/antagonistas & inibidores , Peroxidase/sangue , Pneumonia/patologia , Probabilidade , Distribuição Aleatória , Ratos , Ratos Wistar , Ressuscitação/métodos , Sensibilidade e Especificidade , Choque Hemorrágico/metabolismo
7.
Shock ; 29(2): 181-8, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17693927

RESUMO

We have previously demonstrated that inhibition of the serine-threonine phosphatase PP2A resulted in increased c-jun N-terminal kinase (JNK) activity, and that the regulatory subunit, A/alpha of PP2A, was physically associated with the JNK. Because there exists additional examples of phosphatases serving as negative regulators of multiple members of mitogen-activated protein kinase (MAPK) pathways in Drosophila and yeast, we hypothesized that PP2A may serve a homologous function in mammalian cells affording the regulation of additional upstream kinases in the JNK pathway. In human monocytes, activation of JNK by LPS proceeds through the MAPK kinase kinase MEKK-1 and, subsequently, the MAPK kinases MKK4 and/or MKK7. Using the human monocyte cell line THP-1, we show that pharmacological manipulation of the activity of PP2A seemed to regulate not only JNK but also the upstream kinases MKK4 and MEKK-1. Using coimmunoprecipitation, overexpression of tagged recombinant JNK, and bacterial two-hybrid strategies, evidence for physical interactions between the structural subunit, PP2A-A/alpha and MEKK-1, MKK4, and JNK was observed. These studies suggest that the target of regulation by PP2A includes upstream kinases in the JNK MAPK pathway. Furthermore, PP2A-A/alpha seems to serve as a structural protein to foster protein-protein interactions affording specificity of the regulation among members of this MAP kinase pathway.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Fosfatase 2/metabolismo , Western Blotting , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Humanos , Imunoprecipitação , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/genética , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , MAP Quinase Quinase 7/genética , MAP Quinase Quinase 7/metabolismo , MAP Quinase Quinase Quinase 1/genética , MAP Quinase Quinase Quinase 1/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/genética , Oxazóis/farmacologia , Ligação Proteica , Proteína Fosfatase 2/antagonistas & inibidores , Proteína Fosfatase 2/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Técnicas do Sistema de Duplo-Híbrido
8.
Shock ; 28(5): 554-63, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17589386

RESUMO

Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) is a nuclear receptor that regulates diverse biological functions including inflammation. The PPARgamma ligands have been reported to exert cardioprotective effects and attenuate myocardial reperfusion injury. Here, we examined the molecular mechanisms of their anti-inflammatory effects. Male Wistar rats were subjected to myocardial ischemia and reperfusion and were treated with the PPAR-gamma ligands, 15-deoxy-Delta-prostaglandin J2 (15d-PGJ2) or ciglitazone, or with vehicle only, in the absence or presence of the selective PPAR-gamma antagonist GW-9662. In vehicle-treated rats, myocardial injury was associated with elevated tissue activity of myeloperoxidase, indicating infiltration of neutrophils, and elevated plasma levels of creatine kinase and tumor necrosis factor-alpha. These events were preceded by activation of the nuclear factor-kappaB pathway. The PPAR-gamma DNA binding was also increased in the heart after reperfusion. Treatment with ciglitazone or 15d-PGJ2 reduced myocardial damage and neutrophil infiltration and blunted creatine kinase levels and cytokine production. The beneficial effects of both ligands were associated with enhancement of PPAR-gamma DNA binding and reduction of nuclear factor-kappaB activation. Treatment with 15d-PGJ2, but not ciglitazone, enhanced DNA binding of heat shock factor 1 and upregulated the expression of the cardioprotective heat shock protein 70. Treatment with 15d-PGJ2, but not ciglitazone, also induced a significant increase in nuclear phosphorylation of the prosurvival kinase Akt. The cardioprotection afforded by ciglitazone was attenuated by the PPAR-gamma antagonist GW-9662. In contrast, GW-9662 did not affect the beneficial effects afforded by 15d-PGJ2. Thus, our data suggest that treatment with these chemically unrelated PPAR-gamma ligands results in diverse anti-inflammatory mechanisms.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Hipoglicemiantes/farmacocinética , Fatores Imunológicos/farmacologia , Traumatismo por Reperfusão Miocárdica/metabolismo , NF-kappa B/metabolismo , PPAR gama/agonistas , Prostaglandina D2/análogos & derivados , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Fatores de Transcrição/metabolismo , Anilidas/farmacologia , Animais , Creatina Quinase/sangue , Proteínas de Choque Térmico HSP70/biossíntese , Fatores de Transcrição de Choque Térmico , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Ligantes , Masculino , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/patologia , Infiltração de Neutrófilos/efeitos dos fármacos , PPAR gama/metabolismo , Peroxidase/metabolismo , Prostaglandina D2/farmacologia , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/sangue , Regulação para Cima/efeitos dos fármacos
9.
Shock ; 28(3): 353-9, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17545942

RESUMO

Epigallocatechin-3-gallate (EGCG) is the main polyphenolic flavonoid found in green tea. Recent in vitro studies have suggested that EGCG inhibits activation of the nuclear factor-kappaB (NF-kappaB) pathway. The NF-kappaB is a transcriptional factor required for gene expression of many inflammatory mediators, including the inducible isoform of nitric oxide synthase (NOS2). Excessive NO production by NOS2 is directly linked to the vasoplegia, shock, and mortality associated with sepsis. Accordingly, we hypothesized that EGCG administration would inhibit NOS2 gene expression and thereby improve survival in a rodent model of polymicrobial sepsis. Polymicrobial sepsis was induced in male Sprague-Dawley rats (hemodynamic study) and C57BL6 mice (mortality study) via cecal ligation and double puncture (CL2P). Rodents were treated with either EGCG (10 mg/kg intraperitoneally) or vehicle at 1 and 6 h after CL2P and every 12 h thereafter. In the hemodynamic study, mean arterial blood pressure was monitored for 18 h, and rats were killed at 3, 6, and 18 h after CL2P. In the mortality study, survival was monitored for 72 h after CL2P in mice. In vehicle-treated rodents, CL2P was associated with profound hypotension and greater than 80% mortality rate. Epigallocatechin-3-gallate treatment significantly improved both the hypotension and survival. In vitro experiments further showed that EGCG inhibited activation of NF-kappaB and subsequent NOS2 gene expression in a primary culture of rat aortic smooth muscle cells. Epigallocatechin-3-gallate may therefore represent a potential nutritional supplement or pharmacologic agent in patients with sepsis.


Assuntos
Catequina/análogos & derivados , Sepse/tratamento farmacológico , Animais , Pressão Sanguínea/efeitos dos fármacos , Catequina/uso terapêutico , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/antagonistas & inibidores , Nitratos/sangue , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/metabolismo , Nitritos/sangue , Ratos , Ratos Sprague-Dawley , Sepse/mortalidade , Taxa de Sobrevida
10.
Exp Hematol ; 34(4): 414-23, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16569588

RESUMO

OBJECTIVE: It is well known that nonsteroidal anti-inflammatory drugs (NSAIDs), such as acetylsalicylic acid, ibuprofen, and indomethacin, induce anti-inflammatory effects through inhibition of cyclooxygenase enzyme activity. However, it has also been established that a variety of their anti-inflammatory effects are independent of cyclooxygenase. In the search for alternative modes of action, it was found that NSAIDs share some cellular effects with heat shock treatment. This prompted us to investigate whether NSAIDs modulate production of proinflammatory cytokines by mast cells through the heat shock response. MATERIALS AND METHODS: In mouse mast cells, derived from a culture of bone marrow cells of male BALB/cBy and null HSF-1(-/-) mice, responsiveness to heat shock and NSAIDs was monitored by measuring tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6) production and signaling pathways. RESULTS: In bone marrow-derived mast cells (BMMC), we found that heat shock and a number of NSAIDs induced heat shock protein 70 (HSP70), which was closely paralleled with inhibition of IL-6 and TNF-alpha production. Surprisingly, in BMMC from HSF-1(-/-)mice, heat shock and selected NSAIDs were still able to suppress cytokine production in the absence of HSP70 induction. CONCLUSION: In this article, we provide evidence that inhibition of release of proinflammatory cytokines by NSAIDs and heat shock may be attributed to inhibition of the inhibitory nuclear factor kappaB (NF-kappaB) kinase activity, extracellular signal-regulated kinases 1/2, and p38 pathways, resulting in decreased transcriptional activity of the NF-kappaB pathway.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Células da Medula Óssea/metabolismo , Regulação para Baixo/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/biossíntese , Mastócitos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Células da Medula Óssea/citologia , Células Cultivadas , Proteínas de Ligação a DNA/deficiência , Regulação para Baixo/genética , Ativação Enzimática/efeitos dos fármacos , Fatores de Transcrição de Choque Térmico , Resposta ao Choque Térmico , Interleucina-6/biossíntese , Masculino , Mastócitos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas Quinases/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/deficiência , Fator de Necrose Tumoral alfa/biossíntese
11.
Shock ; 23(1): 80-7, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15614136

RESUMO

Endotoxin tolerance has been characterized as diminished TNF-alpha expression after a second LPS stimulus and is dependent on new protein synthesis. LPS-induced expression of TNF-alpha is partly regulated by the p38 mitogen-activated protein (MAP) kinase, which post-transcriptionally stabilizes TNF-alpha mRNA. The dual-specific phosphatase, MKP-1, has been shown to negatively regulate p38 via dephosphorylation. We hypothesized that MKP-1 expression induced during tolerance regulates TNF-alpha expression by inhibiting p38 activity. To test this hypothesis, tolerance was induced in THP-1 cells, and naive or tolerized cells were rechallenged 18 h later with LPS (1 microg/mL) and TNF-alpha production was measured. Under similar conditions, nuclear proteins were isolated after LPS stimulation and were analyzed for phospho-p38 and MKP-1 by Western blot. Transient overexpression of MKP-1 was achieved using an adenoviral expression strategy and infected cells subsequently treated with LPS for TNF-alpha production and p38 activation. Results showed that LPS tolerance was induced as reflected by decreased TNF-alpha. Induction of LPS hyporesponsiveness could be mimicked by overexpression of MKP-1 but not beta-gal. MKP-1 expression was noted only in LPS-tolerized or Ad-MKP-1 infected cells. In the canonical and Ad-MKP-1-mediated tolerance models, decreased phospho-p38 activity was observed. MKP-1s role in mediating endotoxin tolerance was further confirmed by demonstrating the inability to fully tolerize peritoneal macrophages isolated from MKP-1 null mutant (vs. wild type) mice (24% vs. 72% reductions, respectively). These data demonstrate that the dual specific phosphatase MKP-1 is an important mediator of endotoxin tolerance via p38 regulation.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Endotoxinas/metabolismo , Regulação Enzimológica da Expressão Gênica , Proteínas Imediatamente Precoces/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Adenoviridae/genética , Animais , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Relação Dose-Resposta a Droga , Fosfatase 1 de Especificidade Dupla , Ativação Enzimática , Ensaio de Imunoadsorção Enzimática , Humanos , Proteínas Imediatamente Precoces/metabolismo , Imunoprecipitação , Lipopolissacarídeos/metabolismo , Macrófagos/citologia , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Fosfoproteínas Fosfatases/metabolismo , Proteína Fosfatase 1 , Proteínas Tirosina Fosfatases/metabolismo , Processamento Pós-Transcricional do RNA , RNA Mensageiro/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
12.
FASEB J ; 16(3): 327-42, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11874982

RESUMO

The role of nitric oxide (NO) generated by the inducible NO synthase (iNOS) during myocardial ischemia and reperfusion is not understood. We investigated the role of iNOS during early reperfusion damage induced in genetically deficient iNOS (iNOS-/-) mice and wild-type littermates. In wild-type mice, ischemia (60 min) and reperfusion (60 min) induced an elevation in serum levels of creatine phosphokinase and myocardial injury characterized by the presence of scattered apoptotic myocytes and mild neutrophil infiltration. Northern blot analysis showed increased expression of iNOS, whose activity was markedly elevated after reperfusion. Immunohistochemistry showed staining for nitrotyrosine; Western blot analysis showed elevated expression of heat shock protein 70 (HSP70), a putative cardioprotective mediator. Plasma levels of nitrite and nitrate, tumor necrosis factor alpha (TNF-alpha), interleukin 6 (IL-6), and IL-10 were also increased. These events were preceded by degradation of inhibitor kappaBalpha (IkappaBalpha), activation of IkappaB kinase complex (IKK) and c-Jun-NH2-terminal kinase (JNK), and subsequently activation of nuclear factor-kappaB (NF-kappaB) and activator protein 1 (AP-1) as early as 15 min after reperfusion. In contrast, iNOS-/- mice experienced 35% mortality after reperfusion. The extensive myocardial injury was associated with marked apoptosis and infiltration of neutrophils whereas expression of HSP70 was less pronounced. Nitrotyrosine formation and plasma levels of nitrite and nitrate were undetectable. TNF-alpha and IL-6 were increased and IL-10 was reduced in earlier stages of reperfusion. Activation of IKK and JNK and binding activity of NF-kappaB and AP-1 were significantly reduced. Thus, we conclude that iNOS plays a beneficial role in modulating the early defensive inflammatory response against reperfusion injury through regulation of signal transduction.


Assuntos
Traumatismo por Reperfusão Miocárdica/enzimologia , NF-kappa B/metabolismo , Óxido Nítrico Sintase/fisiologia , Fator de Transcrição AP-1/metabolismo , Tirosina/análogos & derivados , Animais , Apoptose , Cardiotônicos/metabolismo , Citocinas/biossíntese , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Infiltração de Neutrófilos , Óxido Nítrico/fisiologia , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Transdução de Sinais , Tirosina/metabolismo
13.
J Leukoc Biol ; 72(5): 1027-36, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12429726

RESUMO

Adenosine is an endogenous nucleoside that regulates numerous cellular functions including inflammation. Adenosine acts via cell-surface receptors subtyped as A1, A2A, A2B, and A3. The A2A receptor (A2AR) has been linked to anti-inflammatory effects of adenosine. Furthermore, microarray analysis revealed increased A2AR mRNA in lipopolysaccharide (LPS)-stimulated monocytes. We hypothesized that endogenous adenosine inhibited LPS-mediated tumor necrosis factor (TNF) production via A2AR stimulation. Using THP-1 cells, our results demonstrated that LPS increased expression of cellular A2AR and adenosine. A2AR agonism with 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamido adenosine (CGS 21680) after LPS decreased TNF production in a dose- and time-dependent manner, whereas A2AR antagonism significantly increased TNF and blocked the inhibitory effect of CGS 21680. This inhibitory pathway involved A2AR stimulation of cyclic adenosine monophosphate (cAMP) to activate protein kinase A, resulting in phosphorylation of cAMP response element-binding protein (CREB). Phospho-CREB had been shown to inhibit nuclear factor-kappaB transcriptional activity, as was observed with CGS 21680 treatment. Thus, following immune activation with LPS, endogenous adenosine mediates a negative feedback pathway to modulate cytokine expression in THP-1 cells.


Assuntos
Adenosina/análogos & derivados , Citocinas/biossíntese , Regulação para Baixo , Monócitos/imunologia , Receptores Purinérgicos P1/fisiologia , Transdução de Sinais , Adenosina/biossíntese , Adenosina/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Humanos , Cinética , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Fenetilaminas/farmacologia , Antagonistas de Receptores Purinérgicos P1 , RNA Mensageiro/biossíntese , Receptor A2A de Adenosina , Receptores Purinérgicos P1/genética , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/biossíntese
14.
Shock ; 17(2): 127-34, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11837788

RESUMO

Sesquiterpene lactones are extracts of common medicinal Asteracae plants used in folk medicine for their anti-inflammatory activity. Recently, in vitro studies have shown that these compounds may interfere with pro-inflammatory gene regulation. This study examines the effects of parthenolide, a sesquiterpene lactone, in experimental myocardial ischemia and reperfusion. Myocardial injury was induced in rats by 30 min occlusion and 120 min reperfusion of the left coronary artery. Parthenolide (250 or 500 microg/kg) or vehicle (0.05% Tween 80, 1 mL/kg) was administered intraperitoneally 10 min before reperfusion. In vehicle-treated rats, ischemia and reperfusion caused myocardial injury, as evaluated by infarct size, serum levels of creatine phosphokinase and by histological examination. Elevated tissue levels of myeloperoxidase activity were indicative of a significant infiltration of neutrophils. This event paralleled the occurrence of oxidative damage, as evaluated by a marked increase in tissue malondialdehyde levels. These inflammatory events were preceded by activation of the IkappaB kinase complex (IKK) and partial disappearance of inhibitor-kappaBalpha (IkappaBalpha) in the cytosol and translocation of the nuclear factor-kappaB (NF-kappaB) to the nucleus, as early as 15 min after reperfusion. Administration of parthenolide ameliorated myocardial injury, lowered serum creatine phosphokinase activity, and reduced neutrophil infiltration and the subsequent oxidative damage. These beneficial effects were associated with inhibition of IKK activity, enhanced stability of IkappaBalpha, and inhibition of nuclear translocation of NF-kappaB. The results of this study suggest that parthenolide may be beneficial for the treatment of reperfusion-induced myocardial damage by inhibition of the IKK/NF-kappaB pathway.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas I-kappa B , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Sesquiterpenos/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Proteínas de Ligação a DNA/metabolismo , Hemodinâmica , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Inibidor de NF-kappaB alfa , Infiltração de Neutrófilos/efeitos dos fármacos , Ratos , Ratos Wistar , Sesquiterpenos/farmacologia
15.
Shock ; 17(4): 274-9, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11954826

RESUMO

Hyperoxia and tumor necrosis factor-alpha (TNFalpha) are two canonical signals centrally involved in the pathophysiology of acute lung injury. We have attempted to elucidate the effects of these two stimuli on the signal transduction pathways of lung parenchymal cells. In cultured human lung epithelial cells, exposure to hyperoxia alone (95% oxygen) did not affect NF-kappaB activation or degradation of the NF-kappaB inhibitory protein, IkappaB alpha. Stimulation with TNFalpha alone increased NF-kappaB activation within 1 h and induced IkappaB alpha degradation within 0.5 h. After TNFalpha alone, NF-kappaB activation returned to baseline within 2 h and this corresponded with near complete IkappaB alpha resynthesis within 1 h of stimulation. In contrast, simultaneous exposure to hyperoxia and TNFalpha prolonged NF-kappaB activation up to 4 h, and IkappaB alpha degradation up to 2 h after stimulation. Hyperoxia did not affect TNFalpha-mediated resynthesis of IkappaB alpha mRNA. Hyperoxia alone did not induce IkappaB kinase (IKK) activity, but significantly prolonged TNFalpha-mediated activation of IKK activity. Hyperoxia alone did not activate the intercellular adhesion molecule-1 (ICAM-1) promoter, but augmented TNFalpha-mediated activation of the ICAM-1 promoter. These data demonstrate that while hyperoxia alone does not affect activation of NF-kappaB, hyperoxia prolongs TNFalpha-mediated activation of NF-kappaB. The mechanism of this effect involves, in part, prolonged degradation of IkappaB alpha resulting from prolonged activation of IKK.


Assuntos
Hiperóxia/metabolismo , Proteínas I-kappa B , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Expressão Gênica , Humanos , Hiperóxia/genética , Quinase I-kappa B , Molécula 1 de Adesão Intercelular/genética , Pulmão/metabolismo , Lesão Pulmonar , Inibidor de NF-kappaB alfa , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/farmacologia
16.
Shock ; 17(2): 91-7, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11837795

RESUMO

The pulmonary vascular endothelium plays a critical role in lung inflammation. As a result of proinflammatory cytokine expression, adhesion molecules are upregulated on the surface of the endothelial cells. Adhesion molecules facilitate recruitment of leukocytes and thus, have been targeted for potential anti-inflammatory strategies. Prior induction of the stress response through thermal stimulation, or heat shock, alters proinflammatory gene expression by attenuating NF-kappaB signaling. As intercellular adhesion molecule-(ICAM) 1 expression is, in part, NF-kappaB-dependent, we hypothesized that heat shock would inhibit ICAM-1 expression. Heat shocking endothelial cells resulted in heat shock protein (HSP) expression as measured by HSP-70 induction, and decreased TNF-alpha-induced ICAM-1 expression in a manner that appeared to be transcriptionally mediated. Following heat shock, decreased TNF-alpha-induced NF-kappaB activation was observed and was associated with preservation of IkappaB-alpha and a decrease in phosphorylated IkappaB-alpha that correlated to inhibition of I kappa kinase (IKK) activity. Interestingly, exposing respiratory epithelial cells to heat shock, which results in NF-kappaB inhibition, did not affect TNF-induced ICAM-1 expression. We conclude that heat shock decreases endothelial cell ICAM-1 expression via inhibition of IKK activity.


Assuntos
Endotélio Vascular/metabolismo , Resposta ao Choque Térmico , Proteínas I-kappa B/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Proteínas I-kappa B/antagonistas & inibidores , Molécula 1 de Adesão Intercelular/efeitos dos fármacos , Molécula 1 de Adesão Intercelular/genética , NF-kappa B/metabolismo , Transcrição Gênica , Fator de Necrose Tumoral alfa/farmacologia
17.
Inflammation ; 26(5): 233-41, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12238566

RESUMO

Interleukin-8 (IL-8) is a principle neutrophil chemoattractant and activator in humans. There is interest in developing novel pharmacological inhibitors of IL-8 gene expression as a means for modulating inflammation in disease states such as acute lung injury. Herein we determined the effects of epigallocatechin-3-gallate (EGCG), a green tea-derived polyphenol, on tumor necrosis factor-alpha (TNF-alpha)-mediated expression of the IL-8 gene in A549 cells. EGCG inhibited TNF-alpha-mediated IL-8 gene expression in a dose response manner, as measured by ELISA and Northern blot analysis. This effect appears to primarily involve inhibition of IL-8 transcription because EGCG inhibited TNF-alpha-mediated activation of the IL-8 promoter in cells transiently transfected with an IL-8 promoter-luciferase reporter plasmid. In addition, EGCG inhibited TNF-alpha-mediated activation of IkappaB kinase and subsequent activation of the IkappaB alpha/NF-kappaB pathway. We conclude that EGCG is a potent inhibitor of IL-8 gene expression in vitro. The proximal mechanism of this effect involves, in part, inhibition of IkappaB kinase activation.


Assuntos
Catequina/análogos & derivados , Catequina/farmacologia , Flavonoides , Interleucina-8/biossíntese , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Mucosa Respiratória/metabolismo , Chá/química , Antioxidantes/farmacologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Quinase I-kappa B , Interleucina-8/genética , NF-kappa B/metabolismo , Fenóis/farmacologia , Polímeros/farmacologia , RNA Mensageiro/antagonistas & inibidores , Mucosa Respiratória/efeitos dos fármacos , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia
18.
Inflammation ; 26(3): 129-37, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12083419

RESUMO

Previous studies demonstrated important interactions between the heat shock response and the IkappaBalpha/NF-kappaB pathway when these two pathways are induced sequentially. One such interaction involves the ability of heat shock to inhibit subsequent degradation of IkappaBalpha in response to a proinflammatory signal. Herein we investigated the temporal relationship between recovery from heat shock and inhibition of IkappaBalpha degradation, and the proximal mechanisms by which heat shock inhibits degradation of IkappaBalpha in macrophages. In RAW 264.7 murine macrophages, prior heat shock inhibited LPS-mediated IkappaBalpha degradation up to 4 h after recovery from heat shock, and this effect correlated with inhibition of LPS-mediated activation of NF-kappaB. Beyond these recovery periods, heat shock did not inhibit IkappaBalpha degradation. IkappaB kinase (IKK) assays demonstrated that heat shock inhibited LPS-mediated activation of IKK up to 1 h after recovery from heat shock. Heat shock also increased intracellular phosphatase activity, and inhibition of intracellular phosphatase activity partially reversed the ability of heat shock to inhibit both LPS-mediated degradation of IkappaBalpha and LPS-mediated activation of IKK. These data demonstrate that the ability of heat shock to inhibit degradation of IkappaBalpha is dependent on the recovery period between the heat shock stimulus and the proinflammatory stimulus. The mechanism by which heat shock inhibits degradation of IkappaBalpha involves dual modulation of IKK and intracellular phosphatase activity.


Assuntos
Resposta ao Choque Térmico , Proteínas I-kappa B/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , NF-kappa B/metabolismo , Animais , Linhagem Celular , Fatores Imunológicos/farmacologia , Camundongos , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Fatores de Tempo
19.
Shock ; 35(4): 367-74, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20926989

RESUMO

Liver X receptor α (LXRα) is a nuclear transcription factor that regulates lipid metabolism. Recently, it has been shown that activation of LXRα with synthetic ligands has anti-inflammatory effects in atherosclerosis and chemical-induced dermatitis. We investigated the effect of the LXRα agonist, T0901317, on lung inflammation in a rodent model of hemorrhagic shock. Hemorrhagic shock was induced in male rats by withdrawing blood to a goal mean arterial blood pressure of 50 mmHg. Blood pressure was maintained at this level for 3 h, at which point rats were rapidly resuscitated with shed blood. Animals were then treated with T0901317 (50 mg · kg) or vehicle i.p. and sacrificed at 1, 2, and 3 h after resuscitation. Treatment with T0901317 significantly improved the cardiac and stroke volume indices as well as the heart rate of rats during the resuscitation period as compared with vehicle-treated rats. The T0901317-treated animals showed significant improvement in the plasma level of lactate, whereas base deficit and bicarbonate levels both trended toward improvement. The T0901317-treated animals also showed lower levels of plasma cytokines and chemokines monocyte chemoattractant protein 1, macrophage inflammatory protein 1α, TNF-α, KC, and IL-6. Lung injury and neutrophil infiltration were reduced by treatment with T0901317, as evaluated by histology and myeloperoxidase assay. At molecular analysis, treatment with T0901317 increased nuclear LXRα expression and DNA binding while also inhibiting activation of nuclear factor κB, a proinflammatory transcription factor, in the lung. Thus, our data suggest that LXRα is an important modulator of the inflammatory response and lung injury after severe hemorrhagic shock, likely through the inhibition of the nuclear factor κB pathway.


Assuntos
Hidrocarbonetos Fluorados/uso terapêutico , NF-kappa B/metabolismo , Receptores Nucleares Órfãos/metabolismo , Choque Hemorrágico/tratamento farmacológico , Choque Hemorrágico/metabolismo , Sulfonamidas/uso terapêutico , Animais , Bicarbonatos/sangue , Quimiocina CCL2/sangue , Quimiocina CCL3/sangue , Colesterol/sangue , Modelos Animais de Doenças , Interleucina-10/sangue , Interleucina-6/sangue , Ácido Láctico/sangue , Receptores X do Fígado , Masculino , Ratos , Ratos Wistar , Choque Hemorrágico/sangue , Choque Hemorrágico/imunologia , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/sangue
20.
Adv Pharmacol Sci ; 2011: 691928, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21603190

RESUMO

Albumin appears to have proinflammatory effects in vitro. We hypothesized that albumin would induce a state of tolerance to subsequent administration of lipopolysaccharide (LPS) in vitro and in vivo. RAW264.7 and primary peritoneal macrophages were treated with increasing doses of bovine serum albumin (BSA) and harvested for NF-κB luciferase reporter assay or TNF-α ELISA. In separate experiments, RAW264.7 cells were preconditioned with 1 mg/mL BSA for 18 h prior to LPS (10 µg/mL) treatment and harvested for NF-κB luciferase reporter assay or TNF-α ELISA. Finally, C57Bl/6 mice were preconditioned with albumin via intraperitoneal administration 18 h prior to a lethal dose of LPS (60 mg/kg body wt). Blood was collected at 6 h after LPS administration for TNF-α ELISA. Albumin produced a dose-dependent and TLR-4-dependent increase in NF-κB activation and TNF-α gene expression in vitro. Albumin preconditioning abrogated the LPS-mediated increase in NF-κB activation and TNF-α gene expression in vitro and in vivo. The clinical significance of these findings remains to be elucidated.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa