Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros











Intervalo de año de publicación
1.
Eur J Cardiothorac Surg ; 40(2): 352-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21242090

RESUMEN

OBJECTIVE: Insufficient myocardial protection is still a considerable cause for in-hospital mortality in children. The purpose of our study was to investigate underlying the basic mechanisms of cardioplegic cardioprotection during hypothermic and normothermic ischemia in a cardiomyocyte cell culture model. METHODS: We cooled cardiomyocytes to 20°C for 20min; during this time, cardiac arrest was simulated by oxidative damage with 2mM H2O2 and cardioplegic solution, followed by rewarming to 37°C. Later on, we analyzed cardiomyocyte cell morphology (phase-contrast-microscopy), viability (trypan blue staining), inflammation (cyclooxygenase-2 (Cox-2) and phosphorylated-extracellular signal-regulated kinase (pERK) 1/2 expression in Western blot analysis), and expression of Akt survival protein (Western blot technique). RESULTS: Hypothermia increases cell survival of cardiomyocytes after cardioplegic ischemia, as demonstrated in significantly higher cell viability and less cell death in these cells compared with normothermic H2O2-damaged cardiomyocytes. As a possible underlying cellular mechanism, we found that, during cold cardioplegic ischemia, ERK 1/2 enzyme is less phosphorylated than under conditions of normothermic cardioplegic ischemia. This is in line with significantly diminished Cox-2 expression during cold cardioplegic ischemia. Moreover, hypothermic cardioplegia preserved cell survival by upregulation of Akt transcription factor in cardiomyocytes. CONCLUSION: In the present cell culture study, we clearly demonstrated that hypothermia exerts additional protection for cardiomyocytes during cardioplegic ischemia. The understanding of underlying basic mechanisms is evident to improve current techniques of myocardial protection.


Asunto(s)
Frío , Paro Cardíaco Inducido , Isquemia Miocárdica/prevención & control , Miocitos Cardíacos/fisiología , Soluciones Cardiopléjicas/farmacología , Supervivencia Celular/fisiología , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Activación Enzimática/fisiología , Humanos , Peróxido de Hidrógeno/farmacología , Microscopía de Contraste de Fase , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Estrés Oxidativo/fisiología , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo
2.
Cryobiology ; 62(1): 53-61, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21168400

RESUMEN

The purpose of our study was to investigate underlying basic mechanisms of hypothermia-induced cardioprotection during oxidative stress in a cardiomyocyte cell culture model. For hypothermic treatment we cooled H9c2 cardiomyocytes to 20°C, maintained 20min at 20°C during which short-term oxidative damage was inflicted with 2mM H(2)O(2,) followed by rewarming to 37°C. Later on, we analyzed lactate dehydrogenase (LDH), caspase-3 cleavage, reactive oxygen species (ROS), mitochondrial activity, intracellular ATP production, cytoprotective signal molecules as well as DNA damage. Hypothermia decreased H(2)O(2) damage in cardiomyocytes as demonstrated in a lower LDH release, less caspase-3 cleavage and less M30 CytoDeath staining. After rewarming H(2)O(2) damaged cells demonstrated a significantly higher reduction rate of intracellular ROS compared to normothermic H(2)O(2) damaged cardiomyocytes(.) This was in line with a significantly greater mitochondrial dehydrogenase activity and higher intracellular ATP content in cooled and rewarmed cells. Moreover, hypothermia preserved cell viability by up-regulation of the anti-apoptotic protein Bcl-2 and a reduction of p53 phosphorylation. DNA damage, proven by PARP-1 cleavage and H2AX phosphorylation, was significantly reduced by hypothermia. In conclusion, we could demonstrate that hypothermia protects cardiomyocytes during oxidative stress by preventing apoptosis via inhibiting mitochondrial dysfunction and DNA damage.


Asunto(s)
Apoptosis/efectos de los fármacos , Hipotermia Inducida , Mitocondrias/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Daño del ADN/efectos de los fármacos , Histonas/efectos de los fármacos , Histonas/metabolismo , Peróxido de Hidrógeno/farmacología , L-Lactato Deshidrogenasa/efectos de los fármacos , L-Lactato Deshidrogenasa/metabolismo , Mitocondrias/metabolismo , Miocitos Cardíacos/citología , Estrés Oxidativo/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Proteína p53 Supresora de Tumor/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
3.
Vascul Pharmacol ; 51(4): 246-52, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19576293

RESUMEN

To protect immature organ systems during corrective cardiac surgery, patients are cooled to a minimal temperature of 17 degrees C during cardiopulmonary bypass (CPB). However hypothermic CPB triggers the whole body inflammatory response and results in unwanted prolonged inflammation. The present study was designed to clarify the hypothermia and rewarming induced mechanisms and examine interventional pharmacological strategies that could prevent prolonged inflammation. Stimulated primary human umbilical vein endothelial cells (HUVECs) were exposed to a dynamic temperature protocol analogous to clinical settings. Furthermore endothelial cells were pretreated with methylprednisolone and/or tacrolimus as well as with MAPK inhibitors (SB203580, U0126 and SP600125). Cell viability, expression of IL-6 and ERK 1/2, p38 and SAPK/JNK were investigated. Stimulated endothelial cells secreted significantly higher IL-6 protein 2h after rewarming in comparison to normothermic control cells. Moreover, dynamic temperature changes lead to increased MAPK phosphorylation. Only the combined pre-treatment with MP and TAC served to inhibit the IL-6 secretion. As intracellular signalling pathway we could demonstrate that SB203580 as specific p38 inhibitor most effectively down regulated the unwanted IL-6 release after cooling and rewarming. Therefore inhibition of p38 or components of the p38 pathway could be a promising and selective antiinflammatory therapeutic target after hypothermic CPB.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Endotelio Vascular/enzimología , Endotelio Vascular/patología , Hipotermia/patología , Mediadores de Inflamación/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Recalentamiento , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Antiinflamatorios no Esteroideos/farmacología , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Humanos , Hipotermia/tratamiento farmacológico , Hipotermia/enzimología , Mediadores de Inflamación/fisiología , Mediadores de Inflamación/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA