Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Cell Death Differ ; 21(8): 1209-17, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24658400

ABSTRACT

The mitochondrial phosphate carrier (PiC) is critical for ATP synthesis by serving as the primary means for mitochondrial phosphate import across the inner membrane. In addition to its role in energy production, PiC is hypothesized to have a role in cell death as either a component or a regulator of the mitochondrial permeability transition pore (MPTP) complex. Here, we have generated a mouse model with inducible and cardiac-specific deletion of the Slc25a3 gene (PiC protein). Loss of PiC protein did not prevent MPTP opening, suggesting it is not a direct pore-forming component of this complex. However, Slc25a3 deletion in the heart blunted MPTP opening in response to Ca(2+) challenge and led to a greater Ca(2+) uptake capacity. This desensitization of MPTP opening due to loss or reduction in PiC protein attenuated cardiac ischemic-reperfusion injury, as well as partially protected cells in culture from Ca(2+) overload induced death. Intriguingly, deletion of the Slc25a3 gene from the heart long-term resulted in profound hypertrophy with ventricular dilation and depressed cardiac function, all features that reflect the cardiomyopathy observed in humans with mutations in SLC25A3. Together, these results demonstrate that although the PiC is not a direct component of the MPTP, it can regulate its activity, suggesting a novel therapeutic target for reducing necrotic cell death. In addition, mice lacking Slc25a3 in the heart serve as a novel model of metabolic, mitochondrial-driven cardiomyopathy.


Subject(s)
Cardiomyopathies/genetics , Mitochondria, Heart/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Proton-Phosphate Symporters/genetics , Animals , Calcium/metabolism , Cardiomyopathies/metabolism , Gene Deletion , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Permeability Transition Pore , Oxidative Stress/genetics , Proton-Phosphate Symporters/deficiency
2.
Am J Physiol Heart Circ Physiol ; 281(6): H2500-10, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11709417

ABSTRACT

Recent investigations have established a role for the beta II-isoform of protein kinase C (PKC beta II) in the induction of cardiac hypertrophy and failure. Although receptors for activated C kinase (RACKs) have been shown to direct PKC signal transduction, the mechanism through which RACK1, a selective PKC beta II RACK, participates in PKC beta II-mediated cardiac hypertrophy and failure remains undefined. We have previously reported that PKC epsilon activation modulates the expression of RACKs, and that altered epsilon-isoform of PKC (PKC epsilon)-RACK interactions may facilitate the genesis of cardiac phenotypes in mice. Here, we present evidence that high levels of PKC epsilon activity are commensurate with impaired left ventricular function (dP/dt = 6,074 +/- 248 mmHg/s in control vs. 3,784 +/- 269 mmHg/s in transgenic) and significant myocardial hypertrophy. More importantly, we demonstrate that high levels of PKC epsilon activation induce a significant colocalization of PKC beta II with RACK1 (154 +/- 7% of control) and a marked redistribution of PKC beta II to the particulate fraction (17 +/- 2% of total PKC beta II in control mice vs. 49 +/- 5% of total PKC beta II in hypertrophied mice), without compensatory changes of the other eight PKC isoforms present in the mouse heart. This enhanced PKC beta II activation is coupled with increased RACK1 expression and PKC beta II-RACK1 interactions, demonstrating PKC epsilon-induced PKC beta II signaling via a RACK1-dependent mechanism. Taken together with our previous findings regarding enhanced RACK1 expression and PKC epsilon-RACK1 interactions in the setting of cardiac hypertrophy and failure, these results suggest that RACK1 serves as a nexus for at least two isoforms of PKC, the epsilon-isoform and the beta II-isoform, thus coordinating PKC-mediated hypertrophic signaling.


Subject(s)
Heart Failure/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Myocardium/enzymology , Peptides/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Actins/genetics , Animals , Cardiomegaly/metabolism , Gene Expression/physiology , Humans , Jurkat Cells , Mice , Mice, Transgenic , Mutagenesis/physiology , Myocardial Contraction/physiology , Myosin Heavy Chains/genetics , Protein Interaction Mapping , Protein Kinase C beta , Protein Kinase C-epsilon , Receptors for Activated C Kinase , Signal Transduction/physiology
3.
J Mol Cell Cardiol ; 33(11): 1989-2005, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11708843

ABSTRACT

Recent studies suggest that ischemia activates Src and members of the mitogen-activated protein (MAP) kinase superfamily and their downstream effectors, including big MAP kinase 1 (BMK1) and p90 ribosomal S6 kinase (p90RSK). It has also been reported that adenosine is released during ischemia and involved in triggering the protective mechanism of ischemic preconditioning. To assess the roles of Src and adenosine in ischemia-induced MAP kinases activation, we utilized the Src inhibitor PP2 (4-Amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine) and the adenosine receptor antagonist 8-(p-sulfophenyl) theophylline (SPT) in perfused guinea pig hearts. PP2 (1 microm) inhibited ischemia-induced Src, BMK1 and JNK activation but not JAK2 and p38 activation. SPT inhibited ischemia-mediated p38 and JNK activation. These results demonstrate that Src family kinase and adenosine regulate MAP kinases by parallel pathways. Preconditioning significantly improved both recovery of developed pressure and dp/dt in isolated guinea pig hearts. Since the protective effect of preconditioning was blocked by PP2 (1 microm) and SPT (50 microm), we next investigated the regulation of Src, MAP kinases and p90RSK during preconditioning. The activity and time course of ERK1/2 was not changed, but p90RSK activation by reperfusion was completely inhibited by preconditioning. In contrast, the activation by ischemia of Src, BMK1, p38 and JNK was significantly faster in preconditioned hearts. Maximal BMK1 activation by ischemia was also significantly enhanced by preconditioning. These data suggest important roles for Src family kinases and adenosine in mediating preconditioning, and suggest specific roles for individual MAP kinases in preconditioning.


Subject(s)
Adenosine/metabolism , MAP Kinase Signaling System , Myocardial Ischemia , Myocardium/metabolism , Myocardium/pathology , src-Family Kinases/metabolism , Animals , Blotting, Western , Dose-Response Relationship, Drug , Enzyme Activation , Guinea Pigs , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 7 , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/metabolism , Perfusion , Precipitin Tests , Reperfusion Injury/metabolism , Time Factors , p38 Mitogen-Activated Protein Kinases
4.
J Mol Cell Cardiol ; 33(11): 2015-22, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11708845

ABSTRACT

Recent studies reveal that brief ethanol exposure induces cardioprotection against simulated ischemia in cardiomyocytes by the activation of protein kinase C- epsilon. The present study tests the ability of ethanol to induce protection in rabbit hearts in which infarct size was the end-point and explores the signal transduction pathways involved. In isolated rabbit hearts, 50 m m ethanol infused for 5 min with 10 min of washout prior to 30 min of regional ischemia reduced infarct size (triphenyltetrazolium chloride staining) by 49%. Neither adenosine receptor blockade with 8-(p -sulfophenyl) theophylline nor the free radical scavenger N-2-mercaptopropionyl glycine inhibited the protection triggered by ethanol. In contrast, protein kinase C inhibition with chelerythrine, protein tyrosine kinase inhibition with genistein, and blockade of ATP-sensitive potassium channels (K(ATP)) with either 5-hydroxydecanoate or glibenclamide did abolish protection. Thus, transient ethanol exposure followed by washout prior to ischemia elicits a preconditioning-like effect involving protein kinase C, at least one protein tyrosine kinase, and K(ATP)channels, but neither adenosine nor free radicals.


Subject(s)
Alcohols/pharmacology , Ethanol/pharmacology , Heart/drug effects , Ischemic Preconditioning, Myocardial , Myocardial Infarction/prevention & control , Myocardium/metabolism , Adenosine Triphosphate/metabolism , Alkaloids , Analysis of Variance , Animals , Anti-Arrhythmia Agents/pharmacology , Benzophenanthridines , Decanoic Acids/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Genistein/pharmacology , Glyburide/pharmacology , Heart/physiology , Hemodynamics , Hydroxy Acids/pharmacology , Isoenzymes/metabolism , Phenanthridines/pharmacology , Potassium Channels/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase C-epsilon , Rabbits , Rats , Signal Transduction , Time Factors
5.
Basic Res Cardiol ; 96(3): 207-18, 2001.
Article in English | MEDLINE | ID: mdl-11403414

ABSTRACT

In the 15 years since the first publication on ischemic preconditioning (PC), our knowledge of this phenomenon has increased exponentially. While the original studies described the early phase of ischemic PC, we now know that a second or late phase of ischemic PC also exists. In particular, the late phase of PC is triggered by factors such as adenosine, opioids, radicals, and nitric oxide. These factors in turn initiate a molecular chain reaction, which includes activation of serine/threonine kinases, tyrosine kinases, and mitogen-activated protein kinases. Subsequently, this cascade of reactions modulates a plethora of cardioprotective proteins including heat shock proteins, KATP channels, nitric oxide synthase, cyclooxygenase-2, and antioxidants. However, despite this phenomenal amount of information, the construction of a unifying hypothesis describing the signaling mechanism of late PC has proved challenging. The purpose of this article, therefore, is to review the current literature and hypothesis regarding the signaling system in the late phase of ischemic PC, to tackle areas of controversy within this model, and to address potential future directions of investigation that will hopefully promote the generation of a unifying paradigm.


Subject(s)
Ischemic Preconditioning, Myocardial , Myocardial Infarction/prevention & control , Myocardial Infarction/physiopathology , Protein Kinases/physiology , Forecasting , Heat-Shock Proteins/pharmacology , Heat-Shock Proteins/physiology , Humans , Myocardial Infarction/metabolism , Protein Kinase C/pharmacology , Protein Kinase C/physiology , Protein Kinases/pharmacology , Protein-Tyrosine Kinases/pharmacology , Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology
6.
Circ Res ; 88(12): 1306-13, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11420308

ABSTRACT

An essential role for protein kinase C epsilon (PKCepsilon) has been shown in multiple forms of cardioprotection; however, there is a distinct paucity of information concerning the signaling architecture that is responsible for the manifestation of a protective phenotype. We and others have recently shown that signal transduction may proceed via the formation of signaling complexes (Circ Res. 2001;88:59-62). In order to understand if the assembly of multiprotein complexes is the manner by which signaling is conducted in cardioprotection, we designed a series of experiments to characterize the associations of Src tyrosine kinase with PKCepsilon in a conscious rabbit model of nitric oxide (NO)-induced late preconditioning. Our data demonstrate that PKCepsilon and Src can form functional signaling modules in vitro: PKCepsilon interacts with Src; the association with PKCepsilon activates Src; and adult cardiac cells receiving recombinant adenoviruses encoding PKCepsilon exhibit increased Src activity. Furthermore, our results show that NO-induced late preconditioning involved PKCepsilon-Src module formation and enhanced the enzymatic activity of PKCepsilon-associated Src. Inhibition of PKC blocked cardioprotection, module formation, and PKCepsilon-associated Src activity, providing direct evidence for a functional role of the PKCepsilon-Src module in the orchestration of NO-induced cardioprotection in conscious rabbits.


Subject(s)
Ischemic Preconditioning, Myocardial , Isoenzymes/metabolism , Nitric Oxide/metabolism , Protein Kinase C/metabolism , Signal Transduction/physiology , src-Family Kinases/metabolism , Animals , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Glutathione Transferase/genetics , Isoenzymes/genetics , Macromolecular Substances , Mice , Multiprotein Complexes , Myocardium/cytology , Myocardium/metabolism , Nitric Oxide/pharmacology , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Protein Binding/physiology , Protein Kinase C/genetics , Protein Kinase C-epsilon , Rabbits , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Transfection , src-Family Kinases/genetics
7.
J Am Coll Cardiol ; 37(2): 601-7, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11216986

ABSTRACT

OBJECTIVES: Is the timing of exposure critical for ethanol's ability to induce cardioprotection? BACKGROUND: Acute ethanol exposure has been reported to mimic ischemic preconditioning in vitro, but it failed to protect in situ. We hypothesized that these conflicting findings were related to ethanol's presence during ischemia in situ. METHODS: The effect on infarct size (triphenyltetrazolium chloride) of acute ethanol exposure (0.35, 0.7, and 1.4 g/kg IV) 10 min before ischemia was measured in open-chest rabbits after 30 min of regional ischemia and reperfusion and was compared to ethanol's ability to reduce infarct size in isolated hearts in which the timing of ethanol exposure could be varied. RESULTS: Ethanol exposure in situ shortly before ischemia did not reduce infarct size. Moreover, ethanol abolished protection from both ischemic preconditioning and mitochondrial KATP channel activation. In contrast, in buffer-perfused hearts exposed to 10 to 50 mmol/liter ethanol for 5 min followed by washout before ischemia, infarct size was significantly reduced. When ethanol exposure was prolonged until the end of ischemia in isolated hearts, protection was abolished. Conversely, protection was seen when ethanol was infused in situ followed by removal of the heart and perfusion with ethanol-free buffer prior to ischemia in a Langendorff preparation. When 50 min were allowed to metabolize the ethanol prior to ischemia, protection could also be shown in situ. CONCLUSIONS: Ethanol exposure followed by washout or sufficient time to metabolize the alcohol prior to ischemia induces preconditioning-like myocardial protection. However, if present throughout ischemia, ethanol actually blocks all preconditioning-related protection.


Subject(s)
Ethanol/toxicity , Ischemic Preconditioning, Myocardial , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Animals , Ethanol/blood , Female , Male , Myocardium/pathology , Rabbits
8.
Annu Rev Physiol ; 62: 79-109, 2000.
Article in English | MEDLINE | ID: mdl-10845085

ABSTRACT

Ischemic preconditioning is a phenomenon whereby exposure of the myocardium to a brief episode of ischemia and reperfusion markedly reduces tissue necrosis induced by a subsequent prolonged ischemia. It is hoped that elucidation of the mechanism for preconditioning will yield therapeutic strategies capable of reducing myocardial infarction. In the rabbit, the brief period of preconditioning ischemia and reperfusion releases adenosine, bradykinin, opioids, and oxygen radicals. The combined effect of the release of these substances on G proteins and the cell's phospholipases induces the translocation and activation of the epsilon isozyme of protein kinase C. Protein kinase C appears to be the first element of a complex kinase cascade that is activated during the prolonged ischemia in preconditioned hearts. Current evidence indicates that this cascade contains at least one tyrosine kinase and ultimately leads to the activation of p38 mitogen-activated protein kinase. p38 Mitogen-activated protein kinase phosphorylates mitogen-activated protein kinase-activated protein kinase 2. Mitogen-activated protein kinase-activated protein kinase 2 phosphorylates HSP27, a 27-kDa heat shock protein that controls actin filament polymerization, and, therefore, affects the integrity of the cytoskeleton. Finally, mitochondrial adenosine 5'-triphosphate-sensitive K+ channels open, and the latter may be the final mediator of protection for ischemic preconditioning. The protective pathway has many built-in redundancies, perhaps creating a safety factor. These redundancies may also explain some of the species-related differences seen in ischemic preconditioning in which one redundant pathway may predominate over another.


Subject(s)
Ischemic Preconditioning, Myocardial , Potassium Channels/physiology , Receptors, Purinergic P1/physiology , ATP-Binding Cassette Transporters , Animals , Humans , KATP Channels , Myocardium/enzymology , Myocardium/metabolism , Potassium Channels, Inwardly Rectifying , Rabbits
10.
Circ Res ; 86(2): 144-51, 2000 Feb 04.
Article in English | MEDLINE | ID: mdl-10666409

ABSTRACT

Recent studies suggest that p38 mitogen-activated protein kinase (MAPK) may be involved in ischemic preconditioning (PC). To further test this possibility, the regulation of MAPK-activated protein kinase 2 (MAPKAPK2), a kinase immediately downstream from p38 MAPK, and the activity of c-Jun NH(2)-terminal kinase (JNK), a second MAPK, were examined in preconditioned hearts. Isolated, perfused rabbit hearts were subjected to 20 to 30 minutes of global ischemia. Ventricular biopsies before treatment and after 20 minutes of ischemia were homogenized, and the activities of MAPKAPK2 and JNK were evaluated. For the MAPKAPK2 experiments, 7 groups were studied, as follows: control hearts; preconditioned hearts; hearts treated with 500 nmol/L R(-) N(6)-(2-phenylisopropyl) adenosine (PIA), an A(1)-adenosine receptor agonist; preconditioned hearts pretreated with 100 micromol/L 8-(p-sulfophenyl) theophylline (SPT), an adenosine receptor antagonist; preconditioned hearts also treated with SB 203580, a potent inhibitor of p38 MAPK activation; hearts treated with 50 ng/mL anisomycin (a p38 MAPK/JNK activator); and hearts treated with both anisomycin (50 ng/mL) and the tyrosine kinase inhibitor genistein (50 micromol/L). MAPKAPK2 activity was not altered in control hearts after 20 minutes of global ischemia. By contrast, there was a 3.8-fold increase in activity during ischemia in preconditioned hearts. Activation of MAPKAPK2 in preconditioned hearts was blocked by both SPT and SB 203580. MAPKAPK2 activity during ischemia increased 3.5-fold and 3.3-fold in hearts pretreated with PIA or anisomycin, respectively. MAPKAPK2 activation during ischemia in hearts pretreated with anisomycin was blocked by genistein. In separate hearts, anisomycin mimicked the anti-infarct effect of PC, and that protection was abolished by genistein. JNK activity was measured in control and preconditioned hearts. There was a comparable, modest decline in activity during 30 minutes of global ischemia in both groups. As a positive control, a third group of hearts was treated with anisomycin before global ischemia, and in these, JNK activity increased by 290% above baseline. These results confirm that the p38 MAPK/MAPKAPK2 pathway is activated during ischemia only if the heart is in a preconditioned state. These data further support p38 MAPK as an important signaling component in ischemic PC.


Subject(s)
Ischemic Preconditioning, Myocardial , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Myocardial Ischemia/enzymology , Myocardium/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Anisomycin/pharmacology , Coronary Circulation , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Genistein/pharmacology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase 4 , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Kinases/metabolism , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Ischemia/pathology , Protein Synthesis Inhibitors/pharmacology , Rabbits , p38 Mitogen-Activated Protein Kinases
11.
Basic Res Cardiol ; 94(3): 188-98, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10424237

ABSTRACT

Because tyrosine kinase blockade prevents protection by ischemic preconditioning (p.c.) in several species, activation of tyrosine kinase appears to be critical for cardioprotection. The tyrosine kinase's identity, however, is unknown. The present study tested whether activation of a receptor tyrosine kinase, the insulin receptor, could mimic p.c. and if the mechanism of protection was similar to that of p.c. Isolated rabbit hearts were subjected to 30 min of regional ischemia and 2 h of reperfusion. Infarct size was determined by triphenyltetrazolium staining and expressed as a percentage of the area at risk. Infarct size in control hearts was 32.6 +/- 2.3%. A 5-min infusion of insulin (5 mU/ml) followed by a 10-min washout period prior to ischemia significantly reduced infarction to 14.7 +/- 2.1% (P < 0.05). The tyrosine kinase inhibitor genistein (50 microM) given around the insulin infusion blocked protection (28.9 +/- 2.8%). However, when present during the onset of ischemia, genistein had no effect on protection triggered by insulin (14.0 +/- 2.4%; P < 0.05). Inhibition of either PKC by polymyxin B (50 microM) or KATP channels by 5-hydroxydecanoate (100 microM) also failed to prevent protection by insulin (17.5 +/- 3.2% and 17.6 +/- 3.0%, respectively). However, the reduction in infarct size by insulin was significantly attenuated by wortmannin (100 nM), a selective inhibitor of phosphatidylinositol 3-kinase (PI3K, 28.3 +/- 2.2%). Insulin was still able to protect the heart when given only during the reperfusion period (13.2 +/- 3.4%). P.c. reduced infarction to 12.8 +/- 2.0% (P < 0.05) and still offered significant protection in the presence of wortmannin (22.1 +/- 2.4%; P < 0.05). In conclusion, activation of the insulin receptor reduces infarct size in the rabbit heart even when instituted upon reperfusion. However, the mechanism of protection is quite different from that of p.c. and involves activation of PI3K but not PKC or KATP channels.


Subject(s)
Insulin/pharmacology , Myocardial Ischemia/prevention & control , Phosphatidylinositol 3-Kinases/physiology , Potassium Channels/physiology , Protein Kinase C/physiology , Receptor, Insulin/physiology , Animals , Insulin/therapeutic use , Ion Channel Gating , Ischemic Preconditioning, Myocardial , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Rabbits , Signal Transduction
12.
J Cardiovasc Electrophysiol ; 10(5): 741-54, 1999 May.
Article in English | MEDLINE | ID: mdl-10355930

ABSTRACT

Ischemic preconditioning is a phenomenon whereby exposure of the myocardium to a brief episode of ischemia and reperfusion markedly reduces tissue necrosis induced by a subsequent prolonged ischemia. Therefore, it is hoped that elucidation of the mechanism of preconditioning will yield therapeutic strategies capable of reducing myocardial infarction. In the rabbit, the brief period of preconditioning ischemia and reperfusion releases adenosine, bradykinin, opioids, and oxygen radicals that summate to induce the translocation and activation of protein kinase C (PKC). PKC appears to be the first element of a complex kinase cascade that is activated during the prolonged ischemia in preconditioned hearts. Current evidence indicates that PKC activates a tyrosine kinase that leads to the activation of p38 mitogen-activated protein (MAP) kinase or JNK, or possibly both. The stimulation of these stress-activated protein kinases ultimately induces the opening of mitochondrial K(ATP) channels that may be the final mediator of protection by ischemic preconditioning.


Subject(s)
Ischemic Preconditioning, Myocardial , Mitochondria, Heart/enzymology , Potassium Channels/metabolism , Protein Kinases/metabolism , Signal Transduction/physiology , Animals , Biomarkers , Humans , Myocardial Ischemia/enzymology , Myocardial Ischemia/prevention & control , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Rabbits
13.
Am J Physiol ; 276(4): H1361-8, 1999 04.
Article in English | MEDLINE | ID: mdl-10199863

ABSTRACT

Both mitochondrial ATP-sensitive K+ (KATP) channels and the actin cytoskeleton have been proposed to be end-effectors in ischemic preconditioning (PC). For evaluation of the participation of these proposed end effectors, rabbits underwent 30 min of regional ischemia and 3 h of reperfusion. PC by 5-min ischemia + 10-min reperfusion reduced infarct size by 60%. Diazoxide, a mitochondrial KATP-channel opener, administered before ischemia was protective. Protection was lost when diazoxide was given after onset of ischemia. Anisomycin, a p38/JNK activator, reduced infarct size, but protection from both diazoxide and anisomycin was abolished by 5-hydroxydecanoate (5-HD), an inhibitor of mitochondrial KATP channels. Isolated adult rabbit cardiomyocytes were subjected to simulated ischemia by centrifuging the cells into an oxygen-free pellet for 3 h. PC was induced by prior pelleting for 10 min followed by resuspension for 15 min. Osmotic fragility was assessed by adding cells to hypotonic (85 mosmol) Trypan blue. PC delayed the progressive increase in fragility seen in non-PC cells. Incubation with diazoxide or pinacidil was as protective as PC. Anisomycin reduced osmotic fragility, and this was reversed by 5-HD. Interestingly, protection by PC, diazoxide, and pinacidil could be abolished by disruption of the cytoskeleton by cytochalasin D. These data support a role for both mitochondrial KATP channels and cytoskeletal actin in protection by PC.


Subject(s)
Actins/physiology , Adenosine Triphosphate/physiology , Cytoskeleton/physiology , Ischemic Preconditioning, Myocardial , Mitochondria, Heart/metabolism , Mitogen-Activated Protein Kinases , Potassium Channels/physiology , Animals , Anisomycin/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Decanoic Acids/pharmacology , Diazoxide/pharmacology , Enzyme Activation/drug effects , Female , Hydroxy Acids/pharmacology , Male , Osmotic Fragility , Pinacidil/pharmacology , Potassium Channel Blockers , Potassium Channels/drug effects , Rabbits , p38 Mitogen-Activated Protein Kinases
14.
Biochem Cell Biol ; 77(6): 559-67, 1999.
Article in English | MEDLINE | ID: mdl-10668633

ABSTRACT

Protein kinase C (PKC), p38 MAP kinase, and mitogen-activated protein kinase-activated kinases 2 and 3 (MAPKAPK2 and MAPKAPK3) have been implicated in ischemic preconditioning (PC) of the heart to reduce damage following a myocardial infarct. This study examined whether extracellular signal-regulated kinase (Erk) 1, p70 ribosomal S6 kinase (p70 S6K), casein kinase 2 (CK2), and other hsp27 kinases are also activated by PC, and if they are required for protection in rabbit hearts. CK2 and hsp27 kinase activities declined during global ischemia in control hearts, whereas PC with 5 min ischemia and 10 min reperfusion increased their activities during global ischemia. Resource Q chromatography resolved two distinct peaks of hsp27 phosphotransferase activities; the first peak (at 0.36 M NaCl) appeared to correspond to the 55-kDa MAPKAPK2. Erk1 activity was elevated in both control and PC hearts after post-ischemic reperfusion, but no change was observed in p70 S6K activity. Infarct size (measured by triphenyltetrazolium staining) in isolated rabbit hearts subjected to 30 min regional ischemia and 2 h reperfusion was 31.0+/-2.6% of the risk zone in controls and was 10.3+/-2.2% in PC hearts (p<0.001). Neither the CK2 inhibitor 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB) nor the Mek1/2 inhibitor PD98059 infused during ischemia blocked protection by PC. The activation of CK2 and Erk1 in ischemic preconditioned hearts appear to be epiphenomena and not required for the reduction of infarction from myocardial ischemia.


Subject(s)
Ischemic Preconditioning, Myocardial , Myocardial Ischemia/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Casein Kinase II , Chromatography, Ion Exchange , Enzyme Activation , Female , Immunoblotting , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Male , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Myocardial Infarction/enzymology , Rabbits , Ribosomal Protein S6 Kinases/metabolism
15.
Circulation ; 98(9): 899-905, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9738645

ABSTRACT

BACKGROUND: The role of protein phosphatases (PPs) during ischemic preconditioning in the rabbit heart was examined. METHODS AND RESULTS: Fostriecin, a potent inhibitor of PP2A, was administered to isolated rabbit hearts starting either 15 minutes before or 10 minutes after the onset of a 30-minute period of regional ischemia and continuing until the onset of reperfusion. After 2 hours of reperfusion, infarct size was measured with triphenyltetrazolium chloride. In a second study with isolated rabbit cardiomyocytes, the effect of fostriecin pretreatment was assessed by measuring changes in cell osmotic fragility during simulated ischemia. PP1 and PP2A activities of isolated control and ischemically preconditioned cells were also measured. In a third series of experiments, left ventricular biopsies of isolated rabbit hearts were obtained before and at selected times during 60 minutes of global ischemia, and the tissue was assayed for PP1 and PP2A activities. In isolated hearts pretreated with fostriecin, only 8% of the ischemic zone infarcted, significantly less than that in untreated control hearts (33%; P<0.001) but comparable to that in ischemically preconditioned hearts (9%; P<0.001 versus control). Significant protection was also observed in the hearts treated only after the onset of ischemia (18% infarction; P<0.05 versus control). In isolated myocytes, fostriecin also provided protection comparable to that produced by metabolic preconditioning. Preconditioning had no apparent effect on the activity of either PP1 or PP2A in isolated ventricular myocytes or ventricular tissue obtained from heart biopsies. CONCLUSIONS: Fostriecin, a potent inhibitor of PP2A, can protect the rabbit heart from infarction even when administered after the onset of ischemia. But inhibition of either PP1 or PP2A does not appear to be the mechanism of protection from ischemic preconditioning.


Subject(s)
Enzyme Inhibitors/pharmacology , Myocardial Infarction/drug therapy , Myocardial Ischemia/drug therapy , Phosphoprotein Phosphatases/antagonists & inhibitors , Alkenes/pharmacology , Animals , Coronary Circulation , Female , Ischemic Preconditioning, Myocardial , Male , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/pathology , Myocardial Infarction/etiology , Myocardial Infarction/pathology , Myocardial Ischemia/complications , Myocardial Ischemia/pathology , Myocardium/enzymology , Myocardium/pathology , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Polyenes , Protein Phosphatase 2 , Pyrones , Rabbits
16.
J Mol Cell Cardiol ; 30(2): 383-92, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9515015

ABSTRACT

The present study tested the hypothesis that one or more tyrosine kinase(s) are downstream of protein kinase C (PKC) in the signal transduction pathway responsible for the cardioprotective effect of ischemic preconditioning (PC). Isolated rabbit hearts were subjected to 30 min of regional ischemia followed by 2 h of reperfusion. Infarct size was measured by triphenyltetrazolium staining and expressed as a percentage of the area at risk. Infarction in control hearts was 32.9+/-1.8%. Ischemic PC with 5-min ischemia/10-min reperfusion reduced infarct size to 11.5+/-1.5% (P<0.05). Infusion of the tyrosine kinase inhibitors, genistein (50 microM) or lavendustin A (0.5 microM), alone did not affect the level of infarction. When infused around the 5-min PC ischemia genistein failed to block protection (13.7+/-1.0%). However, when present at the onset of the 30-min ischemia both genistein and lavendustin A completely aborted protection (31.4+/-2.0 and 28.1+/-1.5%, respectively). Activation of PKC by phorbol 12-myristate 13-acetate (PMA, 0.05 nmol) was as protective is ischemic PC (14.9+/-3.0%; P<0. 05). Similar to PC, PMA-induced protection was completely prevented by both genistein and lavendustin A. Conversely, anisomycin (50 ng/ml), an activator of MAP kinase kinases (dual tyrosine and threonine kinases), was very protective (7.5+/-1.6%; P<0.05) and this protection was still present when PKC was inhibited by 5 microM chelerythrine (12.1+/-1.6%; P<0.05). In conclusion, activation of a tyrosine kinase during the long ischemia appears to be required for cardioprotection in the rabbit heart. Furthermore, the ability of tyrosine kinase inhibitors to block PMA-induced protection in conjunction with the failure of PKC inhibition to prevent anisomycin-induced protection suggests that the tyrosine kinase is downstream of PKC and that the tyrosine kinase may be a MAP kinase kinase.


Subject(s)
Ischemic Preconditioning, Myocardial , Myocardial Infarction/enzymology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/prevention & control , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Alkaloids , Animals , Anisomycin/pharmacology , Benzophenanthridines , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Female , Genistein/pharmacology , Hemodynamics , Male , Mitogen-Activated Protein Kinase Kinases , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/physiopathology , Phenanthridines/pharmacology , Phenols/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Rabbits , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology
17.
J Mol Cell Cardiol ; 29(1): 207-16, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9040035

ABSTRACT

Previous studies have proposed that oxygen radicals may play a role in the triggering of ischemic preconditioning. However, studies evaluating the effects of radical scavengers have yielded conflicting results, possibly because of differences in the number of preconditioning episodes used. The present study tested whether N-2-mercaptopropionylglycine (MPG) could block protection of both single and multiple episodes of preconditioning in in situ and in vitro rabbit hearts. All hearts were subjected to 30 min of regional ischemia followed by reperfusion for 2 (in vitro) or 3 (in situ) h. Infarct size was measured by tetrazolium. Infarction in control in situ hearts was 37.5+/-3.5% of the risk zone. A single cycle of preconditioning (PC1), with 5 min ischemia/10 min reperfusion, reduced infarct size to 12.3+/-2.0% (P<0.05). Four cycles of preconditioning (PC4) were equally protective. MPG (1 mg/kg/min i.v.) alone had no effect on infarction but abolished protection afforded by PC1 (35.4+/-3.9%). However, MPG failed to block protection in the PC4 group. In isolated control hearts, infarct size was 31.1+/-1.8% and was reduced to 10.2+/-2.2% (P<0.05) by preconditioning. MPG (300 microM) aborted protection. Infusion of hypoxanthine or xanthine oxidase separately in lieu of preconditioning had no effect on infarct size, but induced protection when combined (14.1+/-2.2%; P<0.05). Polymyxin B, an inhibitor of protein kinase C (PKC), abolished this protection (53.1+/-4.1%). In conclusion, oxygen radicals contribute to ischemic preconditioning in the rabbit and appear to do so via activation of PKC. The fact that MPG could not block protection by PC4 suggests that oxygen radicals act in concert with other triggers of preconditioning such as adenosine and bradykinin.


Subject(s)
Free Radical Scavengers/pharmacology , Ischemic Preconditioning, Myocardial , Myocardial Infarction/prevention & control , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , Tiopronin/pharmacology , Analysis of Variance , Animals , Enzyme Activation , Female , Free Radicals , Hemodynamics/drug effects , In Vitro Techniques , Male , Myocardial Infarction/pathology , Rabbits
18.
Br J Pharmacol ; 113(2): 334-5, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7834179

ABSTRACT

The effects of acute intravenous nitroglycerin (NTG) administration on platelet cyclic GMP in relation to changes in indices of preload (end-diastolic volume) and afterload (effective arterial elastance) were evaluated in the anaesthetized mini-pig, using pressure-volume analysis. NTG (1-30 micrograms kg-1 min-1, i.v.) elicited a dose-dependent fall in preload and afterload, and an increase in arterial blood platelet cyclic GMP. Repeated doses of NTG (30 micrograms kg-1 min-1) resulted in tolerance to the preload but not afterload effects. The increases in platelet cyclic GMP were also attenuated, being highly correlated with the preload changes. Therefore, platelet cyclic GMP appears to reflect NTG-induced venous tolerance, rather than arterial responsiveness. The measurement of platelet cyclic GMP may represent a simple approach for monitoring the degree of venous tolerance to NTG in animals or patients, facilitating further mechanistic investigations.


Subject(s)
Blood Platelets/metabolism , Cyclic GMP/blood , Nitroglycerin/pharmacology , Anesthesia , Animals , Blood Platelets/drug effects , Drug Tolerance , Elasticity , Injections, Intravenous , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Nitroglycerin/administration & dosage , Swine , Swine, Miniature , Ventricular Function, Left/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL