Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Immunol ; 187(9): 4778-87, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21957141

RESUMO

Hydrogen sulfide (H(2)S) has been shown to promote transient receptor potential vanilloid type 1 (TRPV1)-mediated neurogenic inflammation in sepsis and its associated multiple organ failure, including acute lung injury (ALI). Accumulating evidence suggests that the cyclooxygenase-2 (COX-2)/PGE(2) pathway plays an important role in augmenting inflammatory immune response in sepsis and respiratory diseases. However, the interactions among H(2)S, COX-2, and PGE(2) in inciting sepsis-evoked ALI remain unknown. Therefore, the aim of this study was to investigate whether H(2)S would upregulate COX-2 and work in conjunction with it to instigate ALI in a murine model of polymicrobial sepsis. Polymicrobial sepsis was induced by cecal ligation and puncture (CLP) in male Swiss mice. dl-propargylglycine, an inhibitor of H(2)S formation, was administrated 1 h before or 1 h after CLP, whereas sodium hydrosulfide, an H(2)S donor, was given during CLP. Mice were treated with TRPV1 antagonist capsazepine 30 min before CLP, followed by assessment of lung COX-2 and PGE(2) metabolite (PGEM) levels. Additionally, septic mice were administrated with parecoxib, a selective COX-2 inhibitor, 20 min post-CLP and subjected to ALI and survival analysis. H(2)S augmented COX-2 and PGEM production in sepsis-evoked ALI by a TRPV1 channel-dependent mechanism. COX-2 inhibition with parecoxib attenuated H(2)S-augmented lung PGEM production, neutrophil infiltration, edema, proinflammatory cytokines, chemokines, and adhesion molecules levels, restored lung histoarchitecture, and protected against CLP-induced lethality. The strong anti-inflammatory and antiseptic actions of selective COX-2 inhibitor may provide a potential therapeutic approach for the management of sepsis and sepsis-associated ALI.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Ciclo-Oxigenase 2/biossíntese , Dinoprostona/biossíntese , Sulfeto de Hidrogênio/farmacologia , Pulmão/patologia , Sepse/metabolismo , Canais de Cátion TRPV/biossíntese , Regulação para Cima/imunologia , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/enzimologia , Animais , Ceco , Ciclo-Oxigenase 2/metabolismo , Ciclo-Oxigenase 2/fisiologia , Dinoprostona/antagonistas & inibidores , Dinoprostona/metabolismo , Ligadura , Pulmão/enzimologia , Pulmão/metabolismo , Masculino , Camundongos , Punções , Sepse/complicações , Sepse/enzimologia , Canais de Cátion TRPV/metabolismo , Regulação para Cima/efeitos dos fármacos
2.
J Immunol ; 185(10): 6265-76, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20926798

RESUMO

Acute lung injury (ALI) is a major cause of mortality in burn patients, even without direct inhalational injury. Identification of early mediators that instigate ALI after burn and of the molecular mechanisms by which they work are of high importance but remain poorly understood. We previously reported that an endogenous neuropeptide, substance P (SP), via binding neurokinin-1 receptor (NK1R), heightens remote ALI early after severe local burn. In this study, we examined the downstream signaling pathway following SP-NK1R coupling that leads to remote ALI after burn. A 30% total body surface area full-thickness burn was induced in male BALB/c wild-type (WT) mice, preprotachykinin-A (PPT-A) gene-deficient mice, which encode for SP, and PPT-A(-/-) mice challenged with exogenous SP. Local burn injury induced excessive SP-NK1R signaling, which activated ERK1/2 and NF-κB, leading to significant upregulation of cyclooxygenase (COX)-2, PGE metabolite, and remote ALI. Notably, lung COX-2 levels were abrogated in burn-injured WT mice by L703606, PD98059, and Bay 11-7082, which are specific NK1R, MEK-1, and NF-κB antagonists, respectively. Additionally, burn-injured PPT-A(-/-) mice showed suppressed lung COX-2 levels, whereas PPT-A(-/-) mice injected with SP showed augmented COX-2 levels postburn, and administration of PD98059 and Bay 11-7082 to burn-injured PPT-A(-/-) mice injected with SP abolished the COX-2 levels. Furthermore, treatment with parecoxib, a selective COX-2 inhibitor, attenuated proinflammatory cytokines, chemokines, and ALI in burn-injured WT mice and PPT-A(-/-) mice injected with SP. To our knowledge, we show for the first time that SP-NK1R signaling markedly elevates COX-2 activity via ERK1/2 and NF-κB, leading to remote ALI after burn.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Queimaduras/metabolismo , Ciclo-Oxigenase 2/metabolismo , Prostaglandinas E/metabolismo , Substância P/metabolismo , Lesão Pulmonar Aguda/imunologia , Animais , Western Blotting , Queimaduras/imunologia , Quimiocinas/biossíntese , Quimiocinas/imunologia , Ciclo-Oxigenase 2/imunologia , Citocinas/biossíntese , Citocinas/imunologia , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/imunologia , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/imunologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/imunologia , NF-kappa B/metabolismo , Prostaglandinas E/imunologia , Transdução de Sinais/imunologia , Substância P/imunologia , Regulação para Cima
3.
Crit Care Med ; 38(2): 619-28, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19851090

RESUMO

OBJECTIVE: To investigate the interaction and involvement of hydrogen sulfide and transient receptor potential vanilloid type 1 in the pathogenesis of sepsis. Hydrogen sulfide has been demonstrated to be involved in many inflammatory states including sepsis. Its contribution in neurogenic inflammation has been suggested in normal airways and urinary bladder. However, whether endogenous hydrogen sulfide would induce transient receptor potential vanilloid type 1-mediated neurogenic inflammation in sepsis remains unknown. DESIGN: Prospective, experimental study. SETTING: Research laboratory. SUBJECT: Male Swiss mice. INTERVENTIONS: Mice were subjected to cecal ligation and puncture-induced sepsis and treated with transient receptor potential vanilloid type 1 antagonist capsazepine (15 mg/kg subcutaneous) 30 mins before cecal ligation and puncture. To investigate hydrogen sulfide-mediated neurogenic inflammation in sepsis, DL-propargylglycine (50 mg/kg intraperitoneal), an inhibitor of hydrogen sulfide formation was administrated 1 hr before or 1 hr after the induction of sepsis, whereas sodium hydrosulfide (10 mg/kg intraperitoneal), a hydrogen sulfide donor, was given at the same time as cecal ligation and puncture. Lung and liver myeloperoxidase activities, liver cystathionine-gamma-lyase activity, plasma hydrogen sulfide level, histopathological examination, and survival studies were determined after induction of sepsis. MEASUREMENTS AND MAIN RESULTS: Capsazepine treatment attenuates significantly systemic inflammation and multiple organ damage caused by sepsis, and protects against sepsis-induced mortality. Similarly, administration of sodium hydrosulfide exacerbates but capsazepine reverses these deleterious effects. In the presence of DL-propargylglycine, capsazepine causes no significant changes to the attenuation of sepsis-associated systemic inflammation, multiple organ damage, and mortality. In addition, capsazepine has no effect on endogenous generation of hydrogen sulfide, suggesting that hydrogen sulfide is located upstream of transient receptor potential vanilloid type 1 activation, and may play a critical role in regulating the production and release of sensory neuropeptides in sepsis. CONCLUSIONS: The present study shows that hydrogen sulfide induces systemic inflammation and multiple organ damage characteristic of sepsis via transient receptor potential vanilloid type 1-mediated neurogenic inflammation.


Assuntos
Sulfeto de Hidrogênio/metabolismo , Inflamação Neurogênica/fisiopatologia , Sepse/metabolismo , Canais de Cátion TRPV/efeitos dos fármacos , Canais de Potencial de Receptor Transitório/efeitos dos fármacos , Alcinos/farmacologia , Animais , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Cistationina gama-Liase/metabolismo , Glicina/análogos & derivados , Glicina/farmacologia , Sulfeto de Hidrogênio/antagonistas & inibidores , Sulfeto de Hidrogênio/sangue , Fígado/metabolismo , Fígado/fisiopatologia , Pulmão/metabolismo , Pulmão/fisiopatologia , Masculino , Camundongos , Insuficiência de Múltiplos Órgãos/metabolismo , Peroxidase/metabolismo , Sepse/fisiopatologia , Sulfetos/farmacologia , Canais de Cátion TRPV/fisiologia , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Canais de Potencial de Receptor Transitório/fisiologia
4.
PLoS One ; 6(9): e24535, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21931742

RESUMO

Hydrogen sulfide (H(2)S) has been shown to induce transient receptor potential vanilloid 1 (TRPV1)-mediated neurogenic inflammation in polymicrobial sepsis. However, endogenous neural factors that modulate this event and the molecular mechanism by which this occurs remain unclear. Therefore, this study tested the hypothesis that whether substance P (SP) is one important neural element that implicates in H(2)S-induced neurogenic inflammation in sepsis in a TRPV1-dependent manner, and if so, whether H(2)S regulates this response through activation of the extracellular signal-regulated kinase-nuclear factor-κB (ERK-NF-κB) pathway. Male Swiss mice were subjected to cecal ligation and puncture (CLP)-induced sepsis and treated with TRPV1 antagonist capsazepine 30 minutes before CLP. DL-propargylglycine (PAG), an inhibitor of H(2)S formation, was administrated 1 hour before or 1 hour after sepsis, whereas sodium hydrosulfide (NaHS), an H(2)S donor, was given at the same time as CLP. Capsazepine significantly attenuated H(2)S-induced SP production, inflammatory cytokines, chemokines, and adhesion molecules levels, and protected against lung and liver dysfunction in sepsis. In the absence of H(2)S, capsazepine caused no significant changes to the PAG-mediated attenuation of lung and plasma SP levels, sepsis-associated systemic inflammatory response and multiple organ dysfunction. In addition, capsazepine greatly inhibited phosphorylation of ERK(1/2) and inhibitory κBα, concurrent with suppression of NF-κB activation even in the presence of NaHS. Furthermore, capsazepine had no effect on PAG-mediated abrogation of these levels in sepsis. Taken together, the present findings show that H(2)S regulates TRPV1-mediated neurogenic inflammation in polymicrobial sepsis through enhancement of SP production and activation of the ERK-NF-κB pathway.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sulfeto de Hidrogênio/farmacologia , NF-kappa B/metabolismo , Sepse/tratamento farmacológico , Sepse/microbiologia , Substância P/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Núcleo Celular/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Inflamação , Masculino , Camundongos , Fosforilação
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa