Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cell Biochem ; 112(10): 2873-81, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21618589

RESUMEN

The cardiotoxicity of doxorubicin limits its clinical use in the treatment of a variety of malignancies. Previous studies suggest that doxorubicin-associated cardiotoxicity is mediated by reactive oxygen species (ROS)-induced apoptosis. We therefore investigated if baicalein, a natural antioxidant component of Scutellaria baicalensis, could attenuate ROS generation and cell death induced by doxorubicin. Using an established chick cardiomyocyte model, doxorubicin (10 µM) increased cell death in a concentration- and time-dependent manner. ROS generation was increased in a dose-response fashion and associated with loss of mitochondrial membrane potential. Doxorubicin also augmented DNA fragmentation and increased the phosphorylation of ROS-sensitive pro-apoptotic kinase c-Jun N-terminal kinase (JNK). Adjunct treatment of baicalein (25 µM) and doxorubicin for 24 h significantly reduced both ROS generation (587 ± 89 a.u. vs. 932 a.u. ± 121 a.u., P < 0.01) and cell death (30.6 ± 5.1% vs. 46.8 ± 8.3%, P < 0.01). The dissipated mitochondrial potential and increased DNA fragmentation were also ameliorated. Along with the reduction of ROS and apoptosis, baicalein attenuated phosphorylation of JNK induced by doxorubicin (1.7 ± 0.3 vs. 3.0 ± 0.4-fold, P < 0.05). Co-treatment of cardiomyocytes with doxorubicin and JNK inhibitor SP600125 (10 µM; 24 h) reduced JNK phosphorylation and enhanced cell survival, suggesting that the baicalein protection against doxorubicin cardiotoxicity was mediated by JNK activation. Importantly, concurrent baicalein treatment did not interfere with the anti-proliferative effects of doxorubicin in human breast cancer MCF-7 cells. In conclusion, baicalein adjunct treatment confers anti-apoptotic protection against doxorubicin-induced cardiotoxicity without compromising its anti-cancer efficacy.


Asunto(s)
Doxorrubicina/farmacología , Flavanonas/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fragmentación del ADN/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Fosforilación/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Scutellaria baicalensis/química
2.
Resuscitation ; 82(1): 115-21, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20951489

RESUMEN

STUDY AIMS: Cardiac arrest mortality is significantly affected by failure to obtain return of spontaneous circulation (ROSC) despite cardiopulmonary resuscitation (CPR). Severe myocardial dysfunction and cardiovascular collapse further affects mortality within hours of initial ROSC. Recent work suggests that enhancement of nitric oxide (NO) signaling within minutes of CPR can improve myocardial function and survival. We studied the role of NO signaling on cardiovascular outcomes following cardiac arrest and resuscitation using endothelial NO synthase knockout (NOS3(-/-)) mice. METHODS: Adult female wild-type (WT) and NOS3(-/-) mice were anesthetized, intubated, and instrumented with left-ventricular pressure-volume catheters. Cardiac arrest was induced with intravenous potassium chloride. CPR was performed after 8min of untreated arrest. ROSC rate, cardiac function, whole-blood nitrosylhemoglobin (HbNO) concentrations, heart NOS3 content and phosphorylation (p-NOS3), cyclic guanosine monophosphate (cGMP), and phospho-troponin I (p-TnI) were measured. RESULTS: Despite equal quality CPR, NOS3(-/-) mice displayed lower rates of ROSC compared to WT (47.6% [10/21] vs. 82.4% [14/17], p<0.005). Among ROSC animals, NOS3(-/-) vs. WT mice exhibited increased left-ventricular dysfunction and 120min mortality. Prior to ROSC, myocardial effectors of NO signaling including cGMP and p-TnI were decreased in NOS3(-/-) vs. WT mice (p<0.05). Following ROSC in WT mice, significant NOS3-dependent increases in circulating HbNO were seen by 120min. Significant increases in cardiac p-NOS3 occurred between end-arrest and 15min post-ROSC, while total NOS3 content was increased by 120min post-ROSC (p<0.05). CONCLUSIONS: Genetic deletion of NOS3 decreases ROSC rate and worsens post-ROSC left-ventricular function. Poor cardiovascular outcomes are associated with differences in NOS3-dependent myocardial cGMP signaling and circulating NO metabolites.


Asunto(s)
ADN/genética , Eliminación de Gen , Paro Cardíaco/genética , Óxido Nítrico Sintasa de Tipo III/genética , Disfunción Ventricular Izquierda/genética , Animales , Western Blotting , Reanimación Cardiopulmonar/métodos , Modelos Animales de Enfermedad , Femenino , Paro Cardíaco/enzimología , Paro Cardíaco/terapia , Ratones , Ratones Endogámicos C57BL , Miocardio/enzimología , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/etiología
3.
Am J Physiol Heart Circ Physiol ; 298(6): H2164-73, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20382860

RESUMEN

Therapeutic hypothermia (TH) is a promising cardioprotective treatment for cardiac arrest and acute myocardial infarction, but its cytoprotective mechanisms remain unknown. In this study, we developed a murine cardiomyocyte model of ischemia-reperfusion injury to better determine the mechanisms of TH cardioprotection. We hypothesized that TH manipulates Akt, a survival kinase that mediates mitochondrial protection by modulating reactive oxygen species (ROS) and nitric oxide (NO) generation. Cardiomyocytes, isolated from 1- to 2-day-old C57BL6/J mice, were exposed to 90 min simulated ischemia and 3 h reperfusion. For TH, cells were cooled to 32 degrees C during the last 20 min of ischemia and the first hour of reperfusion. Cell viability was evaluated by propidium iodide and lactate dehydrogenase release. ROS production was measured by 6-carboxy-2',7'-dichlorodihydrofluorescein diacetate and mitochondrial membrane potential (DeltaPsim) by 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazoly-carbocyanine iodide (JC-1). Phospho (p)-Akt (Thr308), p-Akt (Ser473), and phosphorylated heat shock protein 27 (p-HSP27) (Ser82) were analyzed by Western blot analysis. TH attenuated reperfusion ROS generation, increased NO, maintained DeltaPsim, and decreased cell death [19.3 + or - 3.3% (n = 11) vs. 44.7 + or - 2.7% (n = 10), P < 0.001]. TH also increased p-Akt during ischemia before reperfusion. TH protection and attenuation of ROS were blocked by the inhibition of Akt and NO synthase but not by a cGMP inhibitor. HSP27, a regulator of Akt, also exhibited increased phosphorylation (Ser82) during ischemia with TH. We conclude that TH cardioprotection is mediated by enhanced Akt/HSP27 phosphorylation and enhanced NO generation, resulting in the attenuation of ROS generation and the maintenance of DeltaPsim following ischemia-reperfusion.


Asunto(s)
Hipotermia Inducida/métodos , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas de Choque Térmico HSP27/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/patología , Oxidantes/metabolismo , Fosforilación , Factores de Tiempo
4.
Am J Physiol Heart Circ Physiol ; 298(6): H1761-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20363892

RESUMEN

Therapeutic hypothermia (TH) cardioprotection has recently been associated with increased Akt signaling in a rat model of cardiac arrest. However, it is not known whether Akt is required for this beneficial effect of TH. We used a mouse model of cardiac arrest demonstrating TH cardioprotection to study the response of mice deficient in an Akt1 allele. We hypothesized that Akt1 mediates TH cardioprotection and that decreases in Akt1 content would diminish such protection. Adult C57BL/6 wild-type (WT) mice underwent an 8-min cardiac arrest. After 6 min, the mice were randomized to normothermia (WT(NT), 37 degrees C) or TH (WT(TH), 30 degrees C). Following cardiopulmonary resuscitation and the return of spontaneous circulation (ROSC), the animals were hemodynamically monitored for 240 min (R240). At R240, cardiac tissue Akt content and phosphorylation were assayed. Studies were repeated in Akt1 heterozygous (Akt1(+/-)) mice. As a result, baseline characteristics and ROSC rates were equivalent across groups. At R240, WT(TH) mice exhibited lower heart rate, larger stroke volume, and higher cardiac output than WT(NT) animals (P < 0.05). Cardioprotection in WT(TH) at R240 was associated with increased cardiac Akt phosphorylation at Ser473 and Thr308 compared with that in WT(NT) (P < 0.05). TH-associated alterations in Akt phosphorylation, stroke volume, heart rate, and cardiac output were abrogated in Akt1(+/-) animals. In conclusion, TH improves post-ROSC cardiac function and increases Akt phosphorylation in WT, but not Akt1(+/-), mice. The Akt1 isoform appears necessary for TH-mediated cardioprotection.


Asunto(s)
Gasto Cardíaco/fisiología , Paro Cardíaco/fisiopatología , Hipotermia Inducida/efectos adversos , Proteínas Proto-Oncogénicas c-akt/deficiencia , Proteínas Proto-Oncogénicas c-akt/genética , Animales , Femenino , Paro Cardíaco/complicaciones , Hemodinámica/fisiología , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Volumen Sistólico/fisiología
5.
Am J Respir Cell Mol Biol ; 43(3): 342-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19855087

RESUMEN

Our previous studies revealed that, in a murine model of asthma, mice that received Fas-deficient T cells developed a prolonged phase of airway inflammation, mucus production, and airway hyperreactivity that failed to resolve even 6 weeks after the last challenge. To investigate how Fas-Fas ligand (FasL) interaction occurs between T cells and other cells in vivo, Gld mice with abnormalities of the FasL signaling pathway were used. The reconstituted mice were made by transferring T cells from B6 or Gld mice to Rag(-/-) or FasL-deficient Rag(-/-) mice. We found that Rag(-/-) mice that received B6 T cells resolved the airway inflammation, whereas FasL-deficient Rag(-/-) mice that received Gld T cells developed a prolonged airway inflammation at Day 28, with decreased IFN-gamma production. Both FasL-deficient Rag(-/-) mice that received B6 T cells and Rag(-/-) mice that received Gld T cells also had completely resolved their airway inflammation by Day 28 after challenge. Interestingly, FasL-deficient Rag(-/-) mice that received Gld T cells eventually resolved airway inflammation at Day 42, with a similar level of IFN-gamma production to that of control group. These results demonstrate that FasL expression on either T cells only or non-T cells only was sufficient for the eventual resolution of airway inflammation, and the prolonged airway inflammation in FasL-deficient Rag(-/-) mice that received Gld T cells was correlated with decreased IFN-gamma production by Gld T cells.


Asunto(s)
Asma/prevención & control , Modelos Animales de Enfermedad , Proteína Ligando Fas/fisiología , Sistema Respiratorio/metabolismo , Linfocitos T/metabolismo , Traslado Adoptivo , Animales , Asma/inmunología , Asma/metabolismo , Células Cultivadas , Citocinas/metabolismo , Citometría de Flujo , Proteínas de Homeodominio/fisiología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/trasplante
6.
J Cell Biochem ; 107(4): 697-705, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19388003

RESUMEN

Ischemia/reperfusion (I/R) injury in cardiomyocytes is related to excess reactive oxygen species (ROS) generation and can be modulated by nitric oxide (NO). We have previously shown that grape seed proanthocyanidin extract (GSPE), a naturally occurring antioxidant, decreased ROS and may potentially stimulate NO production. In this study, we investigated whether GSPE administration at reperfusion was associated with cardioprotection and enhanced NO production in a cardiomyocyte I/R model. GSPE attenuated I/R-induced cell death [18.0 +/- 1.8% (GSPE, 50 microg/ml) vs. 42.3 +/- 3.0% (I/R control), P < 0.001], restored contractility (6/6 vs. 0/6, respectively), and increased NO release. The NO synthase (NOS) inhibitor Nomega-nitro-L-arginine methyl ester (L-NAME, 200 microM) significantly reduced GSPE-induced NO release and its associated cardioprotection [32.7 +/- 2.7% (GSPE + L-NAME) vs. 18.0 +/- 1.8% (GSPE alone), P < 0.01]. To determine whether GSPE induced NO production was mediated by the Akt-eNOS pathway, we utilized the Akt inhibitor API-2. API-2 (10 microM) abrogated GSPE-induced protection [44.3% +/- 2.2% (GSPE + API-2) vs. 27.0% +/- 4.3% (GSPE alone), P < 0.01], attenuated the enhanced phosphorylation of Akt at Ser473 in GSPE-treated cells and attenuated GSPE-induced NO increases. Simultaneously blocking NOS activation (L-NAME) and Akt (API-2) resulted in decreased NO levels similar to using each inhibitor independently. These data suggest that in the context of GSPE stimulation, Akt may help activate eNOS, leading to protective levels of NO. GSPE offers an alternative approach to therapeutic cardioprotection against I/R injury and may offer unique opportunities to improve cardiovascular health by enhancing NO production and increasing Akt-eNOS signaling.


Asunto(s)
Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Extractos Vegetales/farmacología , Proantocianidinas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Embrión de Pollo , Extracto de Semillas de Uva , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/metabolismo , Sustancias Protectoras , Semillas , Vitis
7.
Resuscitation ; 77(2): 242-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18096292

RESUMEN

BACKGROUND: Therapeutic hypothermia (TH) represents an important method to attenuate post-resuscitation injury after cardiac arrest. Laboratory investigations have suggested that induction of hypothermia before return of spontaneous circulation (ROSC) may confer the greatest benefit. We hypothesized that a short delay in resuscitation to induce hypothermia before ROSC, even at the expense of more prolonged ischemia, may yield both physiological and survival advantages. METHODS: Cardiac arrest was induced in C57BL/6 mice using intravenous potassium chloride; resuscitation was attempted with CPR and fluid administration. Animals were randomized into three groups (n=15 each): a normothermic control group, in which 8 min of arrest at 37 degrees C was followed by resuscitation; an early intra-arrest hypothermia group, in which 6.5 min of 37 degrees C arrest were followed by 90s of cooling, with resuscitation attempted at 30 degrees C (8 min total ischemia); and a delayed intra-arrest hypothermia group, with 90s cooling begun after 8 min of 37 degrees C ischemia, so that animals underwent resuscitation at 9.5 min. RESULTS: Animals treated with TH demonstrated improved hemodynamic variables and survival compared to normothermic controls. This was the case even when comparing the delayed intra-arrest hypothermia group with prolonged ischemia time against normothermic controls with shorter ischemia time (7-day survival, 4/15 vs. 0/15, p<0.001). CONCLUSIONS: Short resuscitation delays to allow establishment of hypothermia before ROSC appear beneficial to both cardiac function and survival. This finding supports the concept that post-resuscitation injury processes begin immediately after ROSC, and that intra-arrest cooling may serve as a useful therapeutic approach to improve survival.


Asunto(s)
Paro Cardíaco/terapia , Hipotermia Inducida/métodos , Daño por Reperfusión/prevención & control , Análisis de Varianza , Animales , Temperatura Corporal , Reanimación Cardiopulmonar , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Monitoreo Fisiológico/métodos , Distribución Aleatoria , Tasa de Supervivencia
8.
Crit Care Med ; 35(7): 1709-16, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17522572

RESUMEN

OBJECTIVE: Acute changes in tissue CO2 and pH during reperfusion of the ischemic heart may affect ischemia/reperfusion injury. We tested whether gradual vs. acute decreases in CO2 after cardiomyocyte ischemia affect reperfusion oxidants and injury. DESIGN: Comparative laboratory investigation. SETTING: Institutional laboratory. SUBJECTS: Embryonic chick cardiomyocytes. INTERVENTIONS: Microscope fields of approximately 500 chick cardiomyocytes were monitored throughout 1 hr of simulated ischemia (PO2 of 3-5 torr, PCO2 of 144 torr, pH 6.8), followed by 3 hrs of reperfusion (PO2 of 149 torr, PCO2 of 36 torr, pH 7.4), and compared with cells reperfused with relative hypercarbia (PCO2 of 71 torr, pH 6.8) or hypocarbia (PCO2 of 7 torr, pH 7.9). MEASUREMENTS AND MAIN RESULTS: The measured outcomes included cell viability (via propidium iodide) and oxidant generation (reactive oxygen species via 2',7'-dichlorofluorescin oxidation and nitric oxide [NO] via 4,5-diaminofluorescein diacetate oxidation). Compared with normocarbic reperfusion, hypercarbia significantly reduced cell death from 54.8% +/- 4.0% to 26.3% +/- 2.8% (p < .001), significantly decreased reperfusion reactive oxygen species (p < .05), and increased NO at a later phase of reperfusion (p < .01). The NO synthase inhibitor N-nitro-L-arginine methyl ester (200 microM) reversed this oxidant attenuation (p < .05), NO increase (p < .05), and the cardioprotection conferred by hypercarbic reperfusion (increasing death to 54.3% +/- 6.0% [p < .05]). Conversely, hypocarbic reperfusion increased cell death to 80.4% +/- 4.5% (p < .01). It also increased reactive oxygen species by almost two-fold (p = .052), without affecting the NO level thereafter. Increased reactive oxygen species was attenuated by the mitochondrial complex III inhibitor stigmatellin (20 nM) when given at reperfusion (p < .05). Cell death also decreased from 85.9% +/- 4.5% to 52.2% +/- 6.5% (p < .01). The nicotinamide adenine dinucleotide phosphate oxidase inhibitor apocynin (300 microM) had no effect on reperfusion reactive oxygen species. CONCLUSIONS: Altering CO2 content during reperfusion can significantly affect myocardial postresuscitation injury, in part by modifying mitochondrial oxidants and NO synthase-induced NO production.


Asunto(s)
Dióxido de Carbono/sangre , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Oxidantes/metabolismo , Daño por Reperfusión/metabolismo , Animales , Supervivencia Celular , Células Cultivadas , Embrión de Pollo , Inhibidores Enzimáticos/farmacología , Concentración de Iones de Hidrógeno , Hipercapnia/metabolismo , Hipercapnia/fisiopatología , Hipocapnia/metabolismo , Hipocapnia/fisiopatología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Presión Parcial , Polienos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control
9.
Cell Mol Immunol ; 4(1): 43-52, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17349210

RESUMEN

Tumor necrosis factor alpha (TNF-alpha) has been recognized as an activator of nuclear factor kappaB (NF-kappaB), a factor implicated in the protection of many cell types from apoptosis. We and others have presented evidence to suggest that Fas-induced apoptosis may be an important aspect of the resolution of inflammation, and that delayed resolution of inflammation may be directly associated with NF-kappaB-dependent resistance to Fas. Because TNF-alpha activates NF-kappaB in many cell types including inflammatory cells such as eosinophils, we examined effects of TNF-alpha signaling on the Fas-mediated killing of an eosinophilic cell line AML14. While agonist anti-Fas (CH11) treatment induced apoptosis in AML14 cells, no significant cell death occurred in response to TNF-alpha alone. Electrophoretic mobility shift assay (EMSA) revealed that TNF-alpha induced NF-kappaB transactivation in AML14 cells in a time- and dose-dependent fashion, and subsequent supershift assays indicated that the translocated NF-kappaB was the heterodimer p65 (RelA)/p50. Pre-treatment of cells with TNF-alpha dramatically decreased the CH11-induced cell death in a transient fashion, accompanied by suppression of activation of caspase-8 and caspase-3 activation. Inhibition of NF-kappaB transactivation by inhibitors, BAY 11-7085 and parthenolide, reversed the suppression of Fas-mediated apoptosis by TNF-alpha. Furthermore, TNF-alpha up-regulated X-linked inhibitor of apoptosis protein (XIAP) transiently and XIAP levels were correlated with the temporal pattern of TNF-alpha protection against Fas-mediated apoptosis. This finding suggested that TNF-alpha may contribute to the prolonged survival of inflammatory cells by suppression of Fas-mediated apoptosis, the process involved with NF-kappaB transactivation, anti-apoptotic XIAP up-regulation and caspase suppression.


Asunto(s)
Apoptosis/inmunología , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Enfermedad Aguda , Antiinflamatorios no Esteroideos/farmacología , Anticuerpos Monoclonales/farmacología , Apoptosis/genética , Ensayo de Cambio de Movilidad Electroforética , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Proteína Ligando Fas/antagonistas & inhibidores , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/metabolismo , Leucemia Mieloide , FN-kappa B/antagonistas & inhibidores , Nitrilos/farmacología , Sesquiterpenos/farmacología , Sulfonas/farmacología , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacología , Proteína Inhibidora de la Apoptosis Ligada a X/genética
10.
Am J Physiol Heart Circ Physiol ; 292(4): H1995-2003, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17172266

RESUMEN

Optimal timing of therapeutic hypothermia for cardiac ischemia is unknown. Our prior work suggests that ischemia with rapid reperfusion (I/R) in cardiomyocytes can be more damaging than prolonged ischemia alone. Also, these cardiomyocytes demonstrate protein kinase C (PKC) activation and nitric oxide (NO) signaling that confer protection against I/R injury. Thus we hypothesized that hypothermia will protect most using extended ischemia and early reperfusion cooling and is mediated via PKC and NO synthase (NOS). Chick cardiomyocytes were exposed to an established model of 1-h ischemia/3-h reperfusion, and the same field of initially contracting cells was monitored for viability and NO generation. Normothermic I/R resulted in 49.7 +/- 3.4% cell death. Hypothermia induction to 25 degrees C was most protective (14.3 +/- 0.6% death, P < 0.001 vs. I/R control) when instituted during extended ischemia and early reperfusion, compared with induction after reperfusion (22.4 +/- 2.9% death). Protection was completely lost if onset of cooling was delayed by 15 min of reperfusion (45.0 +/- 8.2% death). Extended ischemia/early reperfusion cooling was associated with increased and sustained NO generation at reperfusion and decreased caspase-3 activation. The NOS inhibitor N(omega)-nitro-L-arginine methyl ester (200 microM) reversed these changes and abrogated hypothermia protection. In addition, the PKCepsilon inhibitor myr-PKCepsilon v1-2 (5 microM) also reversed NO production and hypothermia protection. In conclusion, therapeutic hypothermia initiated during extended ischemia/early reperfusion optimally protects cardiomyocytes from I/R injury. Such protection appears to be mediated by increased NO generation via activation of protein kinase Cepsilon; nitric oxide synthase.


Asunto(s)
Hipotermia Inducida , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/citología , Animales , Apoptosis , Caspasa 3/metabolismo , Células Cultivadas , Embrión de Pollo , Pollos , Frío , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Proteína Quinasa C-epsilon/metabolismo
11.
Crit Care Med ; 34(12 Suppl): S474-82, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17114980

RESUMEN

OBJECTIVE: Within minutes of reperfusing ischemic cardiomyocytes, oxidant stress dramatically increases and is associated with postresuscitation injury. Because mitochondria produce deleterious oxidants and useful metabolic substrates, utilization of electron transport chain inhibitors against reperfusion injury, though promising, must not overly compromise recovery of mitochondrial function. This study sought to further characterize the oxidant source at reperfusion and develop a strategy for therapeutic intervention by manipulation of dose, duration, and the degree of reversibility of mitochondrial inhibition. DESIGN: Comparative laboratory investigation. SETTING: Laboratory of a research university. SUBJECTS: Embryonic chick cardiomyocytes. INTERVENTIONS: Synchronously contracting chick cardiomyocytes were exposed to 1 hr of simulated ischemia and 3 hrs of reperfusion and were monitored for cell viability (propidium iodide) and oxidant generation (dichlorofluorescein). Inhibitors were administered either all course or for the first 15 mins of reperfusion. MEASUREMENTS AND MAIN RESULTS: : Application of diethyldithiocarbamic acid, 2-anthracene-carboxylic acid (rhein tech), and alpha-nicotinamide adenine dinucleotide dehydrogenase (NADH) demonstrated attenuation of the oxidant burst. In addition, diethyldithiocarbamic acid (1 mM), rhein tech (0.1 microM), and alpha-NADH (20 microM) significantly attenuated cell death from a control of 49.7% +/- 6.7% to 15.7% +/- 4.7% (n = 5, p < .01), 26.1% +/- 4.1% (n = 5, p < .01), and 13.8% +/- 1.3% (n = 5, p < .001), respectively. All doses of stigmatellin attenuated reactive oxygen species, but only a 2-20 nM dose during the first 15 mins of reperfusion abrogated cell death from 53.8% +/- 3.5% to 10.8% +/- 2.9% (n = 5, p < .001). Increased doses and durations of stigmatellin abolished reactive oxygen species but augmented injury. Although rotenone (5 microM) attenuated reactive oxygen species, no dose or duration of exposure that ameliorated cell death was found. CONCLUSIONS: Early events of reperfusion are marked by rapid mitochondrial oxidant generation and postresuscitation injury. Electron transport chain blockade provides an effective method of attenuating reactive oxygen species. However, inhibitor administration should be both transient and reversible to necessitate cardioprotection and successful metabolic recovery.


Asunto(s)
Mitocondrias Cardíacas/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Animales , Antracenos/farmacología , Ácidos Carboxílicos/farmacología , Reanimación Cardiopulmonar/efectos adversos , Embrión de Pollo , Ditiocarba/farmacología , Relación Dosis-Respuesta a Droga , Transporte de Electrón , Daño por Reperfusión Miocárdica/etiología , Miocitos Cardíacos/metabolismo , NAD/farmacología , Polienos/farmacología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Rotenona/farmacología
12.
Respir Res ; 7: 78, 2006 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-16709253

RESUMEN

BACKGROUND: Effects of respiratory viral infection on airway epithelium include airway hyper-responsiveness and inflammation. Both features may contribute to the development of asthma. Excessive damage and loss of epithelial cells are characteristic in asthma and may result from viral infection. OBJECTIVE: To investigate apoptosis in Adenoviral-infected Guinea pigs and determine the role of death receptor and ligand expression in the airway epithelial response to limit viral infection. METHODS: Animal models included both an Acute and a Chronic Adeno-infection with ovalbumin-induced airway inflammation with/without corticosteroid treatment. Isolated airway epithelial cells were cultured to study viral production after infection under similar conditions. Immunohistochemistry, western blots and viral DNA detection were used to assess apoptosis, death receptor and TRAIL expression and viral release. RESULTS: In vivo and in vitro Adeno-infection demonstrated different apoptotic and death receptors (DR) 4 and 5 expression in response to corticosteroid exposure. In the Acute Adeno-infection model, apoptosis and DR4/5 expression was coordinated and were time-dependent. However, in vitro Acute viral infection in the presence of corticosteroids demonstrated delayed apoptosis and prolonged viral particle production. This reduction in apoptosis in Adeno-infected epithelial cells by corticosteroids exposure induced a prolonged virus production via both DR4 and TRAIL protein suppression. In the Chronic model where animals were ovalbumin-sensitized/challenged and were treated with corticosteroids, apoptosis was reduced relative to adenovirus-infected or corticosteroid alone. CONCLUSION: Our data suggests that apoptosis of infected cells limits viral production and may be mediated by DR4/5 and TRAIL expression. In the Acute model of Adeno-infection, corticosteroid exposure may prolong viral particle production by altering this apoptotic response of the infected cells. This results from decreased DR4 and TRAIL expression. In the Chronic model treated with corticosteroids, a similar decreased apoptosis was observed. This data suggests that DR and TRAIL modulation by corticosteroids may be important in viral infection of airway epithelium. The prolonged virus release in the setting of corticosteroids may result from reduced apoptosis and suppressed DR4/TRAIL expression by the infected cells.


Asunto(s)
Infecciones por Adenoviridae/fisiopatología , Infecciones por Adenoviridae/virología , Adenoviridae/crecimiento & desarrollo , Antiinflamatorios/farmacología , Apoptosis , Budesonida/farmacología , Tráquea/virología , Enfermedad Aguda , Infecciones por Adenoviridae/complicaciones , Infecciones por Adenoviridae/metabolismo , Animales , Células Cultivadas , Enfermedad Crónica , Células Epiteliales/virología , Femenino , Cobayas , Ovalbúmina , Neumonía/inducido químicamente , Neumonía/complicaciones , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/metabolismo , Tráquea/patología , Tráquea/fisiopatología , Virión/fisiología
13.
J Exp Med ; 203(5): 1173-84, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16618792

RESUMEN

Persistent airway inflammation, mucus production, and airway hyperreactivity are the major contributors to the frequency and severity of asthma. Why lung inflammation persists in asthmatics remains unclear. It has been proposed that Fas-mediated apoptosis of inflammatory cells is a fundamental mechanism involved in the resolution of eosinophilic airway inflammation. Because infiltrating eosinophils are highly sensitive to Fas-mediated apoptosis, it has been presumed that direct ligation of Fas on eosinophils is involved. Here, we utilize adoptive transfers of T cells to demonstrate that the delayed resolution of eosinophilia in Fas-deficient mice is a downstream effect of Fas deficiency on T cells, not eosinophils. Interestingly, the mice that received Fas-deficient T cells, but not the controls, developed a persistent phase of inflammation that failed to resolve even 6 wk after the last challenge. This persistent phase correlated with decreased interferon (IFN)gamma production by Fas-deficient T cells and could be reproduced with adoptive transfer of IFNgamma-deficient T cells. These data demonstrate that Fas deficiency on T cells is sufficient for the development of long-term allergic airway disease in mice and implies that deregulation of death receptors such as Fas on human T cells could be an important factor in the development and/or chronic nature of asthma.


Asunto(s)
Asma/inmunología , Eosinófilos/inmunología , Linfocitos T/inmunología , Receptor fas/inmunología , Traslado Adoptivo , Animales , Asma/genética , Asma/patología , Asma/terapia , Enfermedad Crónica , Modelos Animales de Enfermedad , Eosinofilia/genética , Eosinofilia/inmunología , Eosinofilia/patología , Eosinofilia/terapia , Eosinófilos/patología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Inflamación/terapia , Interferón gamma/inmunología , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Noqueados , Linfocitos T/patología , Linfocitos T/trasplante , Factores de Tiempo , Receptor fas/genética
14.
Am J Physiol Heart Circ Physiol ; 286(6): H2280-6, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-14975933

RESUMEN

We recently demonstrated that reperfusion rapidly induces the mitochondrial pathway of apoptosis in chick cardiomyocytes after 1 h of simulated ischemia. Here we tested whether ischemia-reperfusion (I/R)-induced apoptosis could be initiated by caspase-dependent cytochrome c release in this model of cardiomyocyte injury. Fluorometric assays of caspase activity showed little, if any, activation of caspases above baseline levels induced by 1 h of ischemia alone. However, these assays revealed rapid activation of caspase-2, yielding a 2.95 +/- 0.52-fold increase (over ischemia only) within the 1st h of reperfusion, whereas activities of caspases-3, -8, and -9 increased only slightly from their baseline levels. The rapid and prominent activation of caspase-2 suggested that it could be an important initiator caspase in this model, and using specific caspase inhibitors given only at the point of reperfusion, we tested this hypothesis. The caspase-2 inhibitor benzyloxycarbonyl-Val-Asp(Ome)-Val-Ala-Asp(Ome)-CH(2)F was the only caspase inhibitor that significantly inhibited cytochrome c release from mitochondria. This inhibitor also completely blocked activation of caspases-3, -8, and -9. The caspase-3/7 inhibitor transiently and only partially blocked caspase-2 activity and was less effective in blocking the activities of caspases-8 and -9. The caspase-8 inhibitor failed to significantly block caspase-2 or -3, and the caspase-9 inhibitor blocked only caspase-9. Furthermore, the caspase-2 inhibitor protected against I/R-induced cell death, but the caspase-8 inhibitor failed to do so. These data suggest that active caspase-2 initiates cytochrome c release after reperfusion and that it is critical for the I/R-induced apoptosis in this model.


Asunto(s)
Caspasas/metabolismo , Citocromos c/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/enzimología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Apoptosis/fisiología , Caspasa 2 , Caspasa 3 , Caspasa 8 , Caspasa 9 , Inhibidores de Caspasas , Embrión de Pollo , Pollos , Inhibidores de Cisteína Proteinasa/farmacología , Daño por Reperfusión Miocárdica/patología , Oligopéptidos/farmacología
15.
Am J Physiol Heart Circ Physiol ; 284(1): H141-50, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12388298

RESUMEN

Although ischemia-reperfusion (I/R) can initiate apoptosis, the timing and contribution of the mitochondrial/cytochrome c apoptosis death pathway to I/R injury is unclear. We studied the timing of cytochrome c release during I/R and whether subsequent caspase activation contributes to reperfusion injury in confluent chick cardiomyocytes. One-hour simulated ischemia followed by 3-h reperfusion resulted in significant cell death, with most cell death evident during the reperfusion phase and demonstrating mitochondrial cytochrome c release within 5 min after reperfusion. By contrast, cells exposed to prolonged ischemia for 4 h had only marginally increased cell death and no detectable cytochrome c release into the cytosol. Caspase activation could not be detected after ischemia only, but it significantly increased after reperfusion. Caspase inhibitors benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone, Ac-Asp-Gln-Thr-Asp-H, or benzyloxycarbonyl-Leu-Glu (Ome)-His-Asp-(Ome)-fluoromethyl ketone given only at reperfusion significantly attenuated cell death and resulted in return of contraction. Antixoxidants decreased cytochrome c release, nuclear condensation, and cell death. These results suggest that reperfusion oxidants initiate cytochrome c release within minutes, and apoptosis within hours, significant enough to increase cell death and contractile dysfunction.


Asunto(s)
Apoptosis/fisiología , Corazón/fisiopatología , Células Musculares/fisiología , Miocardio/patología , Daño por Reperfusión/fisiopatología , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Caspasa 3 , Caspasa 9 , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Núcleo Celular/ultraestructura , Células Cultivadas , Embrión de Pollo , Grupo Citocromo c/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Isquemia/fisiopatología , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/metabolismo , Miocardio/ultraestructura , Factores de Tiempo
16.
Curr Eye Res ; 24(3): 206-13, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12221529

RESUMEN

PURPOSE: To evaluate whether human fetal retinal pigment epithelial (HFRPE) cells express TRAIL (tumor necrosis factor related apoptosis inducing ligand). The role of TRAIL in HFRPE induced apoptosis was evaluated. METHODS: Pure cultures of HFRPE cells were isolated. The expression of TRAIL protein and mRNA in non-activated and IFN-gamma activated HFRPE cells was evaluated with RT-PCR. The role of TRAIL in HFRPE induced apoptosis was assessed by incubating HFRPE cells with human T-cell leukemia line Jurkat (Jkt) in the presence or absence of neutralizing TRAIL antibodies. Cultures were pulsed with [(3)H]-thymidine to measure Jkt cell proliferation. The role of TRAIL was further examined by western blott evaluating the cleavage of caspases 8 and 10 in Jkt cells after their incubation with HFRPE cells. RESULTS: HFRPE cells expressed TRAIL mRNA. The expression of TRAIL mRNA and protein was up-regulated by IFN-gamma activation. However, anti-TRAIL antibodies were not able to prevent the HFRPE induced suppression of Jkt cell proliferation. The caspases 8 and 10 were also not cleaved in Jkt cells after their incubation with IFN-gamma activated HFRPE cells. CONCLUSIONS: Although HFRPE cells express TRAIL and its expression is upregulated by IFN-gamma activation, TRAIL is not involved in HFRPE induced apoptosis in Jkt cells. Currently the role of TRAIL in HFRPE cells is under investigation.


Asunto(s)
Apoptosis/fisiología , Feto/fisiología , Células Jurkat/fisiología , Epitelio Pigmentado Ocular/embriología , Anticuerpos/inmunología , Proteínas Reguladoras de la Apoptosis , Caspasa 10 , Caspasa 8 , Caspasa 9 , Caspasas/metabolismo , División Celular/efectos de los fármacos , Células Cultivadas , Precursores Enzimáticos/metabolismo , Feto/citología , Humanos , Interferón gamma/farmacología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Epitelio Pigmentado Ocular/efectos de los fármacos , ARN Mensajero/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
17.
J Immunol ; 169(7): 3536-44, 2002 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12244143

RESUMEN

TNF family receptors can lead to the activation of NF-kappaB and this can be a prosurvival signal in some cells. Although activation of NF-kappaB by ligation of Fas (CD95/Apo-1), a member of the TNFR family, has been observed in a few studies, Fas-mediated NF-kappaB activation has not previously been shown to protect cells from apoptosis. We examined the Fas-induced NF-kappaB activation and its antiapoptotic effects in a leukemic eosinophil cell line, AML14.3D10, an AML14 subline resistant to Fas-mediated apoptosis. EMSA and supershift assays showed that agonist anti-Fas (CH11) induced nuclear translocation of NF-kappaB heterodimer p65(RelA)/p50 in these cells in both a time- and dose-dependent fashion. The influence of NF-kappaB on the induction of apoptosis was studied using pharmacological proteasome inhibitors and an inhibitor of IkappaBalpha phosphorylation to block IkappaBalpha dissociation and degradation. These inhibitors at least partially inhibited NF-kappaB activation and augmented CH11-induced cell death. Stable transfection and overexpression of IkappaBalpha in 3D10 cells inhibited CH11-induced NF-kappaB activation and completely abrogated Fas resistance. Increases in caspase-8 and caspase-3 cleavage induced by CH11 and in consequent apoptotic killing were observed in these cells. Furthermore, while Fas-stimulation of resistant control 3D10 cells led to increases in the antiapoptotic proteins cellular inhibitor of apoptosis protein-1 and X-linked inhibitor of apoptosis protein, Fas-induced apoptosis in IkappaBalpha-overexpressing cells led to the down-modulation of both of these proteins, as well as that of the Bcl-2 family protein, Bcl-x(L). These data suggest that the resistance of these leukemic eosinophils to Fas-mediated killing is due to induced NF-kappaB activation.


Asunto(s)
Apoptosis/inmunología , Eosinófilos/inmunología , Proteínas I-kappa B , Leucemia Mieloide Aguda/inmunología , Glicoproteínas de Membrana/inmunología , FN-kappa B/metabolismo , Receptor fas/inmunología , Receptor fas/metabolismo , Anticuerpos Monoclonales/farmacología , Caspasa 3 , Caspasa 8 , Caspasa 9 , Inhibidores de Caspasas , Caspasas/metabolismo , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta Inmunológica , Regulación hacia Abajo/inmunología , Activación Enzimática/inmunología , Eosinófilos/enzimología , Eosinófilos/metabolismo , Proteína Ligando Fas , Humanos , Inmunidad Innata , Leucemia Mieloide Aguda/enzimología , Ligandos , Glicoproteínas de Membrana/metabolismo , Inhibidor NF-kappaB alfa , FN-kappa B/antagonistas & inhibidores , FN-kappa B/biosíntesis , Subunidades de Proteína , Factores de Tiempo , Transfección , Células Tumorales Cultivadas , Regulación hacia Arriba/inmunología
18.
Immunology ; 107(1): 77-85, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12225365

RESUMEN

We examined the role of cell surface clustering of beta2-integrin caused by protein kinase C (PKC)-activated-cPLA2 in adhesion of eosinophilic AML14.3D10 (AML) cells. Phorbol 12-myristate 13-acetate (PMA) caused time- and concentration-dependent adhesion of AML cells to plated bovine serum albumin (BSA), which was blocked by anti-CD11b or anti-CD18 monoclonal antibodies (mAb) directed against beta2-integrin. Inhibition of PKC with Ro-31-8220 or rottlerin blocked PMA-induced cell adhesion in a concentration-dependent fashion. Inhibition of cytosolic phospholipase A2 (cPLA2) with trifluoromethyl ketone or methyl arachidonyl fluorophosphonate also blocked PMA-induced cell adhesion. PMA caused time-dependent p42/44 mitogen-activated protein kinase (MAPK) (ERK) phosphorylation in these cells. U0126, a MAPK/extracellular signal-regulated protein kinase kinase (MEK) inhibitor, at the concentrations that blocked PMA-induced ERK phosphorylation, had no effect on PMA stimulated AML cell adhesion. Neither p38 MAPK nor c-Jun N-terminal kinase (JNK) was phosphorylated by PMA. PMA also caused increased cPLA2 activity, which was inhibited by Ro-31-8220, but not U0126. Confocal immunofluorescence microscopy showed that PMA caused clustering of CD11b on the cell surface, which was blocked by either PKC or cPLA2 inhibition. PMA stimulation also caused up-regulation of CD11b on the AML cell surface. However, this up-regulation was not affected by cPLA2- or PKC-inhibition. Using the mAb, CBRM1/5, we also demonstrated that PMA does not induce the active conformation of CD11b/CD18. Our data indicate that PMA causes AML cell adhesion through beta2-integrin by PKC activation of cPLA2. This pathway is independent of MEK/ERK and does not require change of CD11b/CD18 to its active conformation. We find that avidity caused by integrin surface clustering - rather than conformational change or up-regulation of CD11b/CD18 - causes PMA stimulated adhesion of AML cells.


Asunto(s)
Antígenos CD18/fisiología , Eosinófilos/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Fosfolipasas A/metabolismo , Acetato de Tetradecanoilforbol/análogos & derivados , Antígeno CD11b/metabolismo , Antígenos CD18/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Técnicas de Cultivo de Célula , Citosol/enzimología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Eosinófilos/metabolismo , Humanos , Fosfolipasas A/antagonistas & inhibidores , Fosfolipasas A2 , Fosforilación , Albúmina Sérica Bovina/farmacología , Acetato de Tetradecanoilforbol/farmacología , Células Tumorales Cultivadas , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...