Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Mol Cell Cardiol ; 43(3): 262-71, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17632123

RESUMEN

Although protein kinase C (PKC) plays a key role in ischemic preconditioning (IPC), the actual mechanism of that protection is unknown. We recently found that protection from IPC requires activation of adenosine receptors during early reperfusion. We, therefore, hypothesized that PKC might act to increase the heart's sensitivity to adenosine. IPC limited infarct size in isolated rabbit hearts subjected to 30-min regional ischemia/2-h reperfusion and IPC's protection was blocked by the PKC inhibitor chelerythrine given during early reperfusion revealing involvement of PKC at reperfusion. Similarly chelerythrine infused in the early reperfusion period blocked the increased phosphorylation of the protective kinases Akt and ERK1/2 observed after IPC. Infusing phorbol 12-myristate 13-acetate (PMA), a PKC activator, during early reperfusion mimicked IPC's protection. As expected, the protection triggered by PMA at reperfusion was blocked by chelerythrine, but surprisingly it was also blocked by MRS1754, an adenosine A(2b) receptor-selective antagonist, suggesting that PKC was somehow facilitating signaling from the A(2b) receptors. NECA [5'-(N-ethylcarboxamido) adenosine], a potent but not selective A(2b) receptor agonist, increased phosphorylation of Akt and ERK1/2 in a dose-dependent manner. Pretreating hearts with PMA or brief preconditioning ischemia had no effect on phosphorylation of Akt or ERK1/2 per se but markedly lowered the threshold for NECA to induce their phosphorylation. BAY 60-6583, a highly selective A(2b) agonist, also caused phosphorylation of ERK1/2 and Akt. MRS1754 prevented phosphorylation induced by BAY 60-6583. BAY 60-6583 limited infarct size when given to ischemic hearts at reperfusion. These results suggest that activation of cardiac A(2b) receptors at reperfusion is protective, but because of the very low affinity of the receptors endogenous cardiac adenosine is unable to trigger their signaling. We propose that the key protective event in IPC occurs when PKC increases the heart's sensitivity to adenosine so that endogenous adenosine can activate A(2b)-dependent signaling.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Daño por Reperfusión Miocárdica/prevención & control , Proteína Quinasa C/metabolismo , Receptor de Adenosina A2B/metabolismo , Transducción de Señal , Acetamidas/farmacología , Agonistas del Receptor de Adenosina A2 , Adenosina-5'-(N-etilcarboxamida)/farmacología , Alcaloides/farmacología , Aminopiridinas/farmacología , Animales , Benzofenantridinas/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Perfusión , Fosforilación/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Purinas/farmacología , Conejos , Acetato de Tetradecanoilforbol/farmacología
2.
Basic Res Cardiol ; 102(1): 73-9, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16900442

RESUMEN

Nicorandil, a hybrid of nitrate generator and potassium channel opener, protects ischemic myocardium by opening mitochondrial ATP sensitive potassium (mitoK(ATP)) channels. We recently found that nitric oxide (NO) opened K(ATP) channels in rabbit hearts by a protein kinase G (PKG) mechanism. This study examined whether the NO-donor property of nicorandil also contributes to opening of mitoK(ATP) channels through PKG. MitoK(ATP) channel opening was monitored in adult rabbit cardiomyocytes by measuring reactive oxygen species (ROS) production, an established marker of channel opening. Nicorandil increased ROS production in a dose-dependent manner. The selective mitoK(ATP) channel inhibitor 5-hydroxydecanoate (200 microM) completely blocked ROS production by nicorandil at all doses. The PKG inhibitor 8-bromoguanosine-3',5'-cyclic monophosphorothioate, Rpisomer (Rp-8-Br-cGMPs, 50 microM) shifted the dose-ROS production curve to the right with an increase of the EC(50) from 2.4 x 10(-5) M to 6.9 x 10(-5) M. Rp- 8-Br-cGMPs did not affect the increase in ROS production by the selective mitoK(ATP) channel opener diazoxide while it completely blocked increased ROS production from the NO donor S-nitroso-N-acetylpenicillamine (1 microM). Furthermore ODQ, an antagonist of soluble guanylyl cyclase, blocked nicorandil's ability to increase ROS generation. These results indicate that nicorandil, in addition to its direct effect on the channels, opens mitoK(ATP) channels indirectly via a NO-PKG signaling pathway.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Nicorandil/farmacología , Canales de Potasio/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Células Cultivadas , Guanilato Ciclasa/antagonistas & inhibidores , Miocitos Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Conejos
3.
Basic Res Cardiol ; 101(2): 159-67, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16283591

RESUMEN

Acetylcholine (ACh) and opioid receptor agonists trigger the preconditioned phenotype through sequential activation of the epidermal growth factor (EGF) receptor, phosphatidylinositol 3-kinase (PI3-K), Akt, and nitric oxide synthase (NOS), and opening of mitochondrial (mito) K(ATP) channels with the generation of reactive oxygen species (ROS). Although extracellular signal-regulated kinase (ERK) has recently been reported to be part of this pathway, its location has not been determined. To address this issue, we administered a 5-min pulse of ACh (550 microM) prior to 30 min of ischemia in isolated rabbit hearts. It reduced infarction from 30.4 +/- 2.2% of the risk zone in control hearts to 12.3 +/- 2.8% and co-administration of the MEK, and, therefore, downstream ERK inhibitor U0126 abolished protection (29.1 +/- 4.6% infarction) con.rming ERK's involvement. MitoK(ATP) opening was monitored in adult rabbit cardiomyocytes by measuring ROS production with MitoTracker Red. ROS production was increased by each of three G protein-coupled agonists: ACh (250 microM), bradykinin (BK) (500 nM), and the delta-opioid agonist DADLE (20 nM). Co-incubation with the MEK inhibitors U0126 (500 nM) or PD 98059 (10 microM) blocked the increased ROS production seen with all three agonists. Direct activation of its receptor by EGF increased ROS production and PD 98059 blocked that increase, thus placing ERK downstream of the EGF receptor. Desferoxamine (DFO) which opens mitoK(ATP) through direct activation of NOS also increased ROS. PD 98059 could not block DFO-induced ROS production, placing ERK upstream of NOS. In isolated hearts, ACh caused phosphorylation of both Akt and ERK. U0126 blocked phosphorylation of ERK but not of Akt. The PI3-K inhibitor wortmannin blocked both. Together these data indicate that ERK is located between Akt and NOS.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Corazón/fisiopatología , Precondicionamiento Isquémico Miocárdico , Transducción de Señal/fisiología , Acetilcolina/farmacología , Animales , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Femenino , Corazón/efectos de los fármacos , Corazón/fisiología , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Mitocondrias/metabolismo , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Infarto del Miocardio/fisiopatología , Técnicas de Cultivo de Órganos , Canales de Potasio/metabolismo , Conejos , Especies Reactivas de Oxígeno
4.
Circ Res ; 97(4): 329-36, 2005 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-16037573

RESUMEN

Ischemic and pharmacological preconditioning can be triggered by an intracellular signaling pathway in which Gi-coupled surface receptors activate a cascade including phosphatidylinositol 3-kinase, endothelial nitric oxide synthase, guanylyl cyclase, and protein kinase G (PKG). Activated PKG opens mitochondrial KATP channels (mitoKATP) which increase production of reactive oxygen species. Steps between PKG and mitoKATP opening are unknown. We describe effects of adding purified PKG and cGMP on K+ transport in isolated mitochondria. Light scattering and respiration measurements indicate PKG induces opening of mitoKATP similar to KATP channel openers like diazoxide and cromakalim in heart, liver, and brain mitochondria. This effect was blocked by mitoKATP inhibitors 5-hydroxydecanoate, tetraphenylphosphonium, and glibenclamide, PKG-selective inhibitor KT5823, and protein kinase C (PKC) inhibitors chelerythrine, Ro318220, and PKC-epsilon peptide antagonist epsilonV(1-2). MitoKATP are opened by the PKC activator 12-phorbol 13-myristate acetate. We conclude PKG is the terminal cytosolic component of the trigger pathway; it transmits the cardioprotective signal from cytosol to inner mitochondrial membrane by a pathway that includes PKC-epsilon.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/fisiología , Citosol/metabolismo , Precondicionamiento Isquémico Miocárdico , Mitocondrias Cardíacas/metabolismo , Transducción de Señal/fisiología , Adenosina Trifosfato/farmacología , Animales , Encéfalo/metabolismo , GMP Cíclico/fisiología , Masculino , Mitocondrias Hepáticas/metabolismo , Consumo de Oxígeno , Canales de Potasio/fisiología , Proteína Quinasa C/fisiología , Ratas , Ratas Wistar , Acetato de Tetradecanoilforbol/farmacología , Compuestos de Tetraetilamonio/farmacología
5.
Vascul Pharmacol ; 42(5-6): 201-9, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15922253

RESUMEN

Acetylcholine (ACh) and bradykinin (BK) are potent pharmacological agents which mimic ischemic preconditioning (IPC) enabling hearts to resist infarction during a subsequent period of ischemia. The cardioprotective pathways activated by BK but not ACh may also protect when activated at reperfusion. ACh and BK stimulate Gi/o-linked receptors and ultimately mediate protection by opening mitochondrial ATP-sensitive potassium channels with the generation of reactive oxygen species that act as second messengers to activate protein kinase C (PKC). There appear to be key differences, however, in the pathways prior to potassium channel opening for these two receptors. This review aims to summarize what is currently known about pharmacological preconditioning by ACh and BK with an emphasis on differences that are seen in the signal transduction cascades. Understanding the cellular basis of protection by ACh and BK is a critical step towards developing pharmacological agents that will prevent infarction during ischemia resulting from coronary occlusion or heart attack.


Asunto(s)
Acetilcolina/farmacología , Bradiquinina/farmacología , Precondicionamiento Isquémico Miocárdico , Acetilcolina/uso terapéutico , Animales , Bradiquinina/uso terapéutico , Humanos
6.
Am J Physiol Heart Circ Physiol ; 287(2): H712-8, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15044194

RESUMEN

Exogenous nitric oxide (NO) triggers a preconditioning-like effect in heart via a pathway that is dependent on reactive oxygen species. This study examined the signaling pathway by which the NO donor S-nitroso-N-acetylpenicillamine (SNAP, 2 microM) triggers its anti-infarct effect. Isolated rabbit hearts experienced 30 min of regional ischemia and 120 min of subsequent reperfusion. Infarct size was determined by triphenyltetrazolium chloride staining. Infarct size was reduced from 30.5 +/- 3.0% of the risk zone in control hearts to 10.2 +/- 2.0% in SNAP-treated hearts. Bracketing the SNAP infusion with either the guanylyl cyclase blocker 1H-[1,2,4]oxadiazole[4,3-a]quinoxalin-1-one (2 microM) or the mitochondrial ATP-sensitive K(+) (mitoK(ATP)) channel blocker 5-hydroxydecanoate (200 microM) completely blocked the infarct-sparing effect of SNAP (34.3 +/- 3.8 and 32.2 +/- 1.6% infarction, respectively). Pretreatment of hearts with 8-(4-chlorophenylthio)-guanosine 3',5'-cyclic monophosphate (10 microM), which is a cell-permeable cGMP analog that activates protein kinase G, mimicked the preconditioning effect of SNAP by reducing infarct size to 7.5 +/- 1.1% of the risk zone. This salutary effect was abolished by either the free radical scavenger N-(2-mercaptopropionyl)glycine (1 mM) or 5-hydroxydecanoate (100 microM; 28.9 +/- 2.7 and 33.6 +/- 5.0% infarction of the risk zone, respectively). To confirm these functional data and the effect of SNAP on the guanylyl cyclase-protein kinase G signaling pathway, cGMP levels were measured. SNAP increased the level from 0.18 +/- 0.04 to 0.61 +/- 0.14 pmol/mg of protein (P < 0.05). These data suggest that exogenous NO triggers the preconditioning effect by initiating a cascade of events including stimulation of guanylyl cyclase to make cGMP, activation of protein kinase G, opening of mitoK(ATP) channels, and, finally, production of reactive oxygen species.


Asunto(s)
GMP Cíclico/metabolismo , Corazón/efectos de los fármacos , Precondicionamiento Isquémico Miocárdico , Proteínas de la Membrana/metabolismo , Donantes de Óxido Nítrico/farmacología , S-Nitroso-N-Acetilpenicilamina/farmacología , Animales , Femenino , Ventrículos Cardíacos , Hemodinámica , Técnicas In Vitro , Masculino , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Canales de Potasio , Conejos
7.
Am J Physiol Heart Circ Physiol ; 286(1): H468-76, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12958031

RESUMEN

Bradykinin (BK) mimics ischemic preconditioning by generating reactive oxygen species (ROS). To identify intermediate steps that lead to ROS generation, rabbit cardiomyocytes were incubated in reduced MitoTracker Red stain, which becomes fluorescent after exposure to ROS. Fluorescence intensity in treated cells was expressed as a percentage of that in paired, untreated cells. BK (500 nM) caused a 51 +/- 16% increase in ROS generation (P < 0.001). Coincubation with either the BK B2-receptor blocker HOE-140 (5 microM) or the free radical scavenger N-(2-mercaptopropionyl)glycine (1 mM) prevented this increase, which confirms that the response was receptor mediated and ROS were actually being measured. Closing mitochondrial ATP-sensitive K+ (mitoKATP) channels with 5-hydroxydecanoate (5-HD, 1 mM) prevented increased ROS generation. BK-induced ROS generation was blocked by Nomega-nitro-m-arginine methyl ester (m-NAME, 200 microM), which implicates nitric oxide as an intermediate. Blockade of guanylyl cyclase with 1-H-[1,2,4]oxadiazole[4,3-a]quinoxalin-1-one (ODQ, 10 microM) aborted BK-induced ROS generation but not that from diazoxide, a direct opener of mitoKATP channels. The protein kinase G (PKG) blocker 8-bromoguanosine-3',5'-cyclic monophosphorothioate (25 microM) eliminated the effects of BK. Conversely, direct activation of PKG with 8-(4-chlorophenylthio)-guanosine-3',5'-cyclic monophosphate (100 microM) increased ROS generation (39 +/- 15%; P < 0.004) similar to BK. This increase was blocked by 5-HD. Finally, the nitric oxide donor S-nitroso-N-acetylpenicillamine (1 microM) increased ROS by 34 +/- 6%. This increase was also blocked by 5-HD. In intact rabbit hearts, BK (400 nM) decreased infarction from 30.5 +/- 3.0 of the risk zone in control hearts to 11.9 +/- 1.4% (P < 0.01). This protection was aborted by either 200 microM m-NAME or 2 microM ODQ (35.4 +/- 5.7 and 30.4 +/- 3.0% infarction, respectively; P = not significant vs. control). Hence, BK preconditions through receptor-mediated production of nitric oxide, which activates guanylyl cyclase. The resulting cGMP activates PKG, which opens mitoKATP. Subsequent release of ROS triggers cardioprotection.


Asunto(s)
Bradiquinina/farmacología , Cardiotónicos/farmacología , Glicina/análogos & derivados , Mitocondrias Cardíacas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Adenosina Trifosfato/fisiología , Animales , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Depuradores de Radicales Libres/farmacología , Glicina/farmacología , Guanilato Ciclasa/metabolismo , Técnicas In Vitro , Óxido Nítrico/metabolismo , Canales de Potasio/metabolismo , Conejos , Receptores de Bradiquinina/metabolismo , Compuestos de Sulfhidrilo/farmacología
8.
Exp Hematol ; 31(11): 1089-96, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14585374

RESUMEN

OBJECTIVE: Our goal was to determine the role of p38 mitogen-activated protein kinase (MAPK) signaling in fetal hemoglobin (HbF) induction. Two histone deacetylase inhibitors (HDAIs), sodium butyrate (NB), and trichostatin (TSA) and hemin were analyzed. In addition, the effect of direct activation of p38 MAPK on gamma-globin gene activity was studied. METHOD: Primary erythroid progenitors derived from peripheral blood mononuclear cell and K562 erythroleukemia cells were analyzed. Cells were grown in NB, TSA, hemin, or anisomycin either alone or in the presence of the p38 MAPK inhibitor SB203580. The effects of the various treatments on gamma-globin RNA, HbF, and phosphorylated p38 MAPK levels were measured by RNase protection assay, alkaline denaturation, and Western blot analysis, respectively. A K562 stable line overexpressing constitutively active p38 MAPK was established using MAPK kinase kinase 3 (MKK3) and MKK6, the immediate upstream activators of p38. The direct effect of p38 MAPK overexpression on gamma-globin mRNA synthesis was analyzed. RESULTS: NB and TSA activated p38 MAPK and increased gamma-globin mRNA levels in K562 cells and primary erythroid progenitors. Pretreatment with SB203580 blocked p38 MAPK and gamma-globin gene activation. In contrast, no change in p38 activity was observed with hemin inductions. Direct activation of p38 by anisomycin or constitutive overexpression also increased gamma-globin mRNA in the absence of HbF inducers in wild-type K562 cells and in the MKK stable lines. CONCLUSION: This study supports a novel role for p38 MAPK in gamma-globin regulation in human erythroid progenitors.


Asunto(s)
Células Precursoras Eritroides/metabolismo , Regulación de la Expresión Génica , Globinas/genética , Proteínas Quinasas Activadas por Mitógenos/fisiología , Butiratos/farmacología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Hemoglobina Fetal/genética , Inhibidores de Histona Desacetilasas , Humanos , Ácidos Hidroxámicos/farmacología , Células K562 , Activación Transcripcional , Proteínas Quinasas p38 Activadas por Mitógenos
9.
J Mol Cell Cardiol ; 35(6): 653-60, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12788383

RESUMEN

Acetylcholine (ACh), like ischemic preconditioning (PC), protects against infarction and is dependent on generation of reactive oxygen species (ROS). To investigate the mechanism by which ACh causes ROS production, isolated adult rabbit cardiomyocytes underwent a timed incubation in reduced MitoTracker Red, which is oxidized to a fluorescent form after exposure to ROS. The mitochondrial ATP-sensitive potassium (mK(ATP)) channel opener diazoxide (50 microM) increased fluorescence by 47 +/- 9% (P = 0.007), indicating that opening of mK(ATP) leads to ROS generation, and that increase was blocked by the mK(ATP) blocker 5-hydroxydecanoate (5HD, 1 mM); 250 microM ACh caused a similar increase in ROS generation (+45 +/- 6% for all experiments, P < 0.001). ACh-induced ROS production was prevented by (1) blockade of muscarinic surface receptors with 100 microM atropine (-6 +/- 2%, P = n.s.) or 250 nM 4-DAMP (+5 +/- 13%, P = n.s.), indicating that ACh's effect was receptor mediated; (2) closing K(ATP) channels with either the non-selective channel closer glibenclamide (50 microM) (-1.2 +/- 17%, P = n.s.) or the selective mK(ATP) closer 5HD (-1.8 +/- 9%, P = n.s.), indicating that increased ROS production involved opening of mK(ATP); (3) blockade of mitochondrial electron transport chain with 200 nM myxothiazol (-4 +/- 9%, P = n.s.), indicating ROS came from the mitochondria; (4) addition of 100 nM wortmannin (-13 +/- 12%, P = n.s.), indicating that phosphatidylinositol 3-(PI3)-kinase was involved; and (5) blockade of Src-kinase with 1 microM PP2 (-2 +/- 5%, P = n.s.), indicating the involvement of an Src-kinase. These results support the hypothesis that occupation of muscarinic surface receptors by ACh causes activation of PI3- and Src-kinases that then open mK(ATP) resulting in mitochondrial ROS generation and triggering of the preconditioned state.


Asunto(s)
Acetilcolina/farmacología , Mitocondrias/metabolismo , Células Musculares/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Canales de Potasio/metabolismo , Familia-src Quinasas/metabolismo , Acetilcolina/metabolismo , Animales , Células Cultivadas , Diazóxido/farmacología , Dinitrofenoles/farmacología , Transporte de Electrón , Inhibidores Enzimáticos/farmacología , Radicales Libres , Ventrículos Cardíacos/metabolismo , Precondicionamiento Isquémico , Metacrilatos , Conejos , Especies Reactivas de Oxígeno , Receptores Muscarínicos/metabolismo , Espectrometría de Fluorescencia , Tiazoles/farmacología , Vitamina K 3/farmacología
10.
Basic Res Cardiol ; 97(5): 365-73, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12200636

RESUMEN

Recent evidence suggests that opening of mitochondrial K(ATP) channels in cardiac muscle triggers the preconditioning phenomenon through free radical production. The present study tested the effects of K(ATP) channel openers in a vascular smooth muscle cell model using the fluorescent probe MitoTracker (MTR) Red trade mark for detection of reactive oxygen species (ROS). Rat aortic smooth muscle cells (A7r5) were incubated with 1 micro M reduced MTR (non-fluorescent) and the MTR oxidation product (fluorescent) was quantified. Thirty-minute pretreatment with either diazoxide (200 micro M) or pinacidil (100 micro M), both potent mitochondrial K(ATP) channel openers, increased fluorescent intensity (FI) to 149 and 162 % of control (p < 0.05 for both), respectively, and the K(ATP) channel inhibitor 5-hydroxydecanoate (5 HD) blocked it. Valinomycin, a potassium-selective ionophore, raised FI to 156 % of control (p <: 0.05). However, 5 HD did not affect the valinomycin-induced increase in FI. Inhibition of mitochondrial electron transport (myxothiazol) or uncoupling of oxidative phosphorylation (dinitrophenol) also blocked either valinomycin- or diazoxide-induced increase in FI, and free radical scavengers prevented any diazoxide-mediated increase in fluorescence. Finally the diazoxide-induced increase in fluorescence was not blocked by the PKC inhibitor chelerythrine, but was by HMR 1883, a putative surface K(ATP) channel blocker. Thus opening of K(ATP) channels increases generation of ROS via the mitochondrial electron transport chain in vascular smooth muscle cells. Furthermore, a potassium-selective ionophore can mimic the effect of putative mitochondrial KATP channel openers. We conclude that potassium movement through KATP directly leads to ROS production by the mitochondria.


Asunto(s)
Activación del Canal Iónico/fisiología , Músculo Liso Vascular/fisiología , Canales de Potasio/fisiología , Vasodilatadores/farmacología , Transportadoras de Casetes de Unión a ATP , Animales , Células Cultivadas , Diazóxido/farmacología , Radicales Libres/metabolismo , Canales KATP , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Músculo Liso Vascular/efectos de los fármacos , Pinacidilo/farmacología , Canales de Potasio de Rectificación Interna , Proteína Quinasa C/metabolismo , Especies Reactivas de Oxígeno/metabolismo
11.
Cardiovasc Res ; 55(3): 681-9, 2002 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12160965

RESUMEN

OBJECTIVES: Ischemic preconditioning (PC) reduces myocardial infarction by a mechanism that involves opening of mitochondrial ATP-dependent potassium channels (mK(ATP)), reactive oxygen species (ROS), and possibly activation of p38 mitogen-activated protein kinase (p38 MAPK). The actual order of these steps, however, is a matter of current debate. This study examined whether protection afforded by menadione, which protects by causing mitochondria to produce ROS, requires mK(ATP) opening. In addition, we tested whether protection from anisomycin, a p38 MAPK activator, is dependent on ROS production. METHODS AND RESULTS: Isolated, buffer-perfused rat hearts were pretreated with menadione, and infarction was assessed after 30 min of regional ischemia and 120 min of reperfusion. Menadione reduced infarction in a dose-dependent manner with an EC(50) of 270 nM. Menadione's infarct-limiting effect was insensitive to 200 microM 5-hydroxydecanoate (5HD), an mK(ATP) channel blocker, whereas protection by diazoxide and PC were blocked by 5HD. Anisomycin caused hearts to resist infarction and this protective effect was abrogated by SB203580, a p38 MAPK inhibitor, and 2-mercaptopropionylglycine (MPG), a free radical scavenger. CONCLUSIONS: These results indicate that mK(ATP) opening occurs upstream of mitochondrial ROS generation in the protective pathway. Furthermore, protection afforded by anisomycin was p38 MAPK- and ROS-dependent.


Asunto(s)
Precondicionamiento Isquémico Miocárdico/métodos , Mitocondrias Cardíacas/metabolismo , Infarto del Miocardio/prevención & control , Canales de Potasio/metabolismo , Transducción de Señal/fisiología , Animales , Anisomicina/farmacología , Ácidos Decanoicos/farmacología , Diazóxido/farmacología , Activadores de Enzimas/farmacología , Inhibidores Enzimáticos/farmacología , Depuradores de Radicales Libres/farmacología , Radicales Libres/metabolismo , Hidroxiácidos/farmacología , Imidazoles/farmacología , Masculino , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Infarto del Miocardio/metabolismo , Perfusión , Bloqueadores de los Canales de Potasio/farmacología , Piridinas/farmacología , Ratas , Ratas Wistar , Tiopronina/farmacología , Vitamina K 3/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...