Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Am J Physiol Heart Circ Physiol ; 298(2): H679-87, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20008276

ABSTRACT

Soluble epoxide hydrolase (sEH) metabolizes epoxyeicosatrienoic acids (EETs), primarily 14,15-EET. EETs are derived from arachidonic acid via P-450 epoxygenases and are cardioprotective. We tested the hypothesis that sEH deficiency and pharmacological inhibition elicit tolerance to ischemia via EET-mediated STAT3 signaling in vitro and in vivo. In addition, the relevance of single nucleotide polymorphisms (SNPs) of EPHX2 (the gene encoding sEH) on tolerance to oxygen and glucose deprivation and reoxygenation and glucose repletion (OGD/RGR) was assessed in male C57BL\6J (WT) or sEH knockout (sEHKO) cardiomyocytes by using transactivator of transcription (TAT)-mediated transduction with sEH mutant proteins. Cell death and hydrolase activity was lower in Arg287Gln EPHX2 mutants vs. nontransduced controls. Excess 14,15-EET and SEH inhibition did not improve cell survival in Arg287Gln mutants. In WT cells, the putative EET receptor antagonist, 14,15-EEZE, abolished the effect of 14,15-EET and sEH inhibition. Cotreatment with 14,15-EET and SEH inhibition did not provide increased protection. In vitro, STAT3 inhibition blocked 14,15-EET cytoprotection, but not the effect of SEH inhibition. However, STAT3 small interfering RNA (siRNA) abolished cytoprotection by 14,15-EET and sEH inhibition, but cells pretreated with JAK2 siRNA remained protected. In vivo, STAT3 inhibition abolished 14,15-EET-mediated infarct size reduction. In summary, the Arg287Gln mutation is associated with improved tolerance against ischemia in vitro, and inhibition of sEH preserves cardiomyocyte viability following OGD/RGR via an EET-dependent mechanism. In vivo and in vitro, 14,15-EET-mediated protection is mediated in part by STAT3.


Subject(s)
Epoxide Hydrolases/antagonists & inhibitors , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/metabolism , Animals , Cell Survival/physiology , Cells, Cultured , Disease Models, Animal , Epoxide Hydrolases/genetics , Epoxide Hydrolases/metabolism , Janus Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Polymorphism, Single Nucleotide/genetics
2.
Am J Physiol Heart Circ Physiol ; 295(5): H2128-34, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18835921

ABSTRACT

Soluble epoxide hydrolase (sEH) metabolizes epoxyeicosatrienoic acids (EETs) to dihydroxyeicosatrienoic acids. EETs are formed from arachidonic acid during myocardial ischemia and play a protective role against ischemic cell death. Deletion of sEH has been shown to be protective against myocardial ischemia in the isolated heart preparation. We tested the hypothesis that sEH inactivation by targeted gene deletion or pharmacological inhibition reduces infarct size (I) after regional myocardial ischemia-reperfusion injury in vivo. Male C57BL\6J wild-type or sEH knockout mice were subjected to 40 min of left coronary artery (LCA) occlusion and 2 h of reperfusion. Wild-type mice were injected intraperitoneally with 12-(3-adamantan-1-yl-ureido)-dodecanoic acid butyl ester (AUDA-BE), a sEH inhibitor, 30 min before LCA occlusion or during ischemia 10 min before reperfusion. 14,15-EET, the main substrate for sEH, was administered intravenously 15 min before LCA occlusion or during ischemia 5 min before reperfusion. The EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (EEZE) was given intravenously 15 min before reperfusion. Area at risk (AAR) and I were assessed using fluorescent microspheres and triphenyltetrazolium chloride, and I was expressed as I/AAR. I was significantly reduced in animals treated with AUDA-BE or 14,15-EET, independent of the time of administration. The cardioprotective effect of AUDA-BE was abolished by the EET antagonist 14,15-EEZE. Immunohistochemistry revealed abundant sEH protein expression in left ventricular tissue. Strategies to increase 14,15-EET, including sEH inactivation, may represent a novel therapeutic approach for cardioprotection against myocardial ischemia-reperfusion injury.


Subject(s)
Adamantane/analogs & derivatives , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Epoxide Hydrolases/deficiency , Gene Deletion , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Urea/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/administration & dosage , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/metabolism , Adamantane/administration & dosage , Adamantane/pharmacology , Animals , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Epoxide Hydrolases/genetics , Female , Heart Ventricles/drug effects , Heart Ventricles/enzymology , Injections, Intraperitoneal , Injections, Intravenous , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/enzymology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Time Factors , Urea/administration & dosage , Urea/pharmacology
3.
Am J Physiol Heart Circ Physiol ; 295(1): H409-15, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18502904

ABSTRACT

There is evidence for differences in the response to the treatment of cardiovascular disease in men and women. In addition, there are conflicting results regarding the effectiveness of pharmacologically induced protection or ischemic preconditioning in females. We investigated whether the ability of Met(5)-enkephalin (ME) to reduce cell death after oxygen-glucose deprivation (OGD) is influenced by the presence of 17beta-estradiol (E(2)) in a nitric oxide (NO)- and estrogen receptor-dependent manner. On postnatal day 7 to 8, murine cardiomyocytes from wild-type or inducible NO synthase (iNOS) knockout mice were separated by sex, isolated by collagenase digestion, cultured for 24 h, and subjected to 90 min OGD and 180 min reoxygenation at 37 degrees C (n = 4 to 5 replicates). Cell cultures were incubated in E(2) for 15 min or 24 h before OGD. ME was used to increase cell survival. Cell death was assessed by propidium iodide. More than 300 cells were examined for each treatment. Data are presented as means +/- SE. As a result, in both sexes, ME-induced cell survival was lost in the presence of E(2), and the ability of ME to improve cell survival was restored after treatment with the estrogen receptor antagonist ICI-182780. Furthermore, iNOS was necessary for ME to increase cell survival following OGD in vitro. We conclude that ME-induced reduction in cell death is abolished by E(2) in a sex-independent manner via activation of estrogen receptors, and this interaction is dependent on iNOS.


Subject(s)
Enkephalin, Methionine/metabolism , Estradiol/metabolism , Glucose/deficiency , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Receptors, Opioid, delta/metabolism , Animals , Animals, Newborn , Cell Death , Cell Hypoxia , Cell Survival , Cells, Cultured , Cytoprotection , Enzyme Inhibitors/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Fulvestrant , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Receptors, Opioid, delta/agonists , Sex Factors
4.
Am J Physiol Heart Circ Physiol ; 294(1): H99-H106, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17951370

ABSTRACT

The balance between norepinephrine (NE) synthesis, release, and reuptake is disrupted after acute myocardial infarction, resulting in elevated extracellular NE. Stimulation of sympathetic neurons in vitro increases NE synthesis and the synthetic enzyme tyrosine hydroxylase (TH) to a greater extent than it increases NE reuptake and the NE transporter (NET), which removes NE from the extracellular space. We used TGR(ASrAOGEN) transgenic rats, which lack postinfarct sympathetic hyperactivity, to test the hypothesis that increased cardiac sympathetic nerve activity accounts for the imbalance in TH and NET expression in these neurons after myocardial infarction. TH and NET mRNA levels were identical in the stellate ganglia of unoperated TGR(ASrAOGEN) rats compared with Sprague Dawley (SD) controls, but the threefold increase in TH and twofold increase in NET mRNA seen in the stellate ganglia of SD rats 1 wk after ischemia-reperfusion was absent in TGR(ASrAOGEN) rats. Similarly, the increase in TH and NET protein observed in the base of the SD ventricle was absent in the base of the TGR (ASrAOGEN) ventricle. Neuronal TH content was depleted in the left ventricle of both genotypes, whereas NET was unchanged. Basal heart rate and cardiac function were similar in both genotypes, but TGR(ASrAOGEN) hearts were more sensitive to the beta-agonist dobutamine. Tyramine-induced release of endogenous NE generated similar changes in ventricular pressure and contractility in both genotypes, but postinfarct relaxation was enhanced in TGR(ASrAOGEN) hearts. These data support the hypothesis that postinfarct sympathetic hyperactivity is the major stimulus increasing TH and NET expression in cardiac neurons.


Subject(s)
Heart/innervation , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Sympathetic Nervous System/metabolism , Tyrosine 3-Monooxygenase/metabolism , Adrenergic beta-Agonists/pharmacology , Angiotensinogen/deficiency , Angiotensinogen/genetics , Angiotensinogen/metabolism , Animals , Animals, Genetically Modified , Coronary Vessels/surgery , Disease Models, Animal , Dobutamine/pharmacology , Female , Heart/drug effects , Heart Rate , Ligation , Male , Myocardial Contraction , Myocardial Infarction/enzymology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/physiopathology , Norepinephrine Plasma Membrane Transport Proteins/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Stellate Ganglion/enzymology , Stellate Ganglion/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/enzymology , Sympathetic Nervous System/physiopathology , Sympathomimetics/pharmacology , Tyramine/pharmacology , Tyrosine 3-Monooxygenase/genetics , Up-Regulation , Ventricular Function, Left
5.
Am J Physiol Heart Circ Physiol ; 294(1): H302-10, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17982014

ABSTRACT

Met(5)-enkephalin (ME)-induced cardioprotection occurs via epidermal growth factor receptor (EGFR) transactivation with the subsequent activation of phosphatidylinositol 3-kinase (PI3K). In the present study, we investigated whether there is a sex difference in ME-elicited PI3K signaling. Neonatal murine cardiomyocytes were isolated by collagenase digestion and subjected to 90 min hypoxia and 180 min reoxygenation at 37 degrees C (n = 5 to 7 replicates). PI3K/Akt signaling was interrogated using pharmacological inhibitors and small interfering RNA (siRNA). Cell death was assessed by propidium iodide. More than 300 cells were examined for each treatment. The data are presented as means +/- SE. There was not a sex difference in the basal content of total Akt. ME (100 microM) elicited comparable protection in both sexes. Wortmannin and the nonselective Akt inhibitor IV completely abolished ME-induced protection in male cardiomyocytes but only attenuated protection in female cardiomyocytes. Isoform-selective knockdown of Akt in males with siRNAs against Akt1/2 completely abolished ME-induced cardioprotection, whereas the siRNAs against Akt3 only attenuated protection of approximately 40%. In contrast, in females the siRNAs against Akt1/2 attenuated and against Akt3 eliminated ME-induced cardioprotection. There is not a sex difference in the degree of ME-induced protection, and there is a sex difference in the cardioprotective signaling pathways after the administration of ME; ME-induced cardioprotection in males primarily utilizes a PI3K/Akt1/2 pathway and in females primarily utilizes a PI3K/Akt3 pathway. The incomplete loss of protection in females following the blockade of PI3K suggests that additional factors may facilitate the maintenance or function of activated Akt.


Subject(s)
Enkephalin, Methionine/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Androstadienes/pharmacology , Animals , Animals, Newborn , Cell Survival , Cells, Cultured , Chromones/pharmacology , Female , Male , Mice , Morpholines/pharmacology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/enzymology , Myocardium/pathology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Time Factors , Wortmannin
6.
Cytokine ; 36(1-2): 9-16, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17150369

ABSTRACT

Cardiotrophin-1 (CT-1) was identified as a growth factor for cardiac myocytes and CT-1 protects myocytes from cell death. Adult CT-1(-/-) mice exhibit neural deficits including the loss of preganglionic sympathetic neurons, but their autonomic and cardiac parameters have not been examined. We used these mice to determine if the absence of CT-1 or loss of preganglionic sympathetic input altered heart rate, left ventricular pressure, cardiac contractility (dP/dt), or cell death following ischemia-reperfusion. Basal heart rate was increased in CT-1(-/-) mice, and this difference was abolished by ganglionic block. Left ventricular pressure and dP/dt were unchanged. Dobutamine stimulated similar increases in heart rate and dP/dt in both genotypes, but ventricular pressure was significantly lower in CT-1 nulls. Cardiac expression of interleukin-6 (IL-6) mRNA was increased significantly in CT-1 null mice, while leukemia inhibitory factor (LIF) mRNA was unchanged. Infarct size normalized to area at risk was no different in CT-1(-/-) mice (33.8+/-1.0% vs. 37.7+/-3.2% WT) 24h after ischemia-reperfusion. Induction of IL-6 mRNA after infarct was significantly abrogated in CT-1 null mice compared to wild-type mice, but LIF mRNA-induction remained significant in CT-1 null mice and might contribute to cardiac protection in the absence of CT-1.


Subject(s)
Cytokines/deficiency , Cytokines/metabolism , Gene Expression , Interleukin-6/genetics , Leukemia Inhibitory Factor/genetics , Animals , Body Weight , Cytokines/genetics , Gene Expression Regulation , Heart Injuries/genetics , Heart Injuries/metabolism , Heart Injuries/pathology , Heart Injuries/physiopathology , Heart Rate , Mice , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Organ Size , RNA, Messenger/genetics
7.
Am J Physiol Heart Circ Physiol ; 288(4): H1955-64, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15563540

ABSTRACT

Our previous studies indicated that opioid-induced cardioprotection occurs via activation of mitochondrial ATP-sensitive K(+) (K(ATP)) channels. However, other elements of the Met(5)-enkephalin (ME) cardioprotection pathway are not fully characterized. In the present study, we investigated the role of tyrosine kinase, MAPK, and phosphatidylinositol 3-kinase (PI3K) signaling in ME-induced protection. Ca(2+)-tolerant, adult rabbit cardiomyocytes were isolated by collagenase digestion and subjected to simulated ischemia for 180 min. ME was administered 15 min before the 180 min of simulated ischemia; blockers were administered 15 min before ME. Cell death was assessed by trypan blue as a function of time. The epidermal growth factor receptor (EGFR) kinase inhibitor AG-1478 (250 nM) blocked ME-induced protection, but the inactive analog AG-9 (100 microM) did not. Treatment with herbimycin (1 microM) completely eliminated ME-induced protection. To verify that ME activates EGFR and to determine the involvement of Src, Western blotting of EGFR was performed after ME administration with and without herbimycin A. ME resulted in herbimycin-sensitive robust phosphorylation of EGFR at Tyr(992) and Tyr(1068). Administration of the selective MAPK inhibitor PD-98059 (10 nM) and the specific MEK1/2 inhibitor U-0126 (10 microM) also inhibited ME-induced cardioprotection. ME-induced ERK1/2 phosphorylation was significantly reduced by PD-98059, the EGFR kinase inhibitor PD-153035 (10 microM), and chelerythrine (2 microM). The PI3K inhibitor LY-294002 (20 microM) abrogated ME-induced protection, and ME-induced Akt phosphorylation at Ser(473) was suppressed by LY-294002, PD-153035, and chelerythrine. We conclude that ME-induced cardioprotection is mediated via Src-dependent EGFR transactivation and activation of the PI3K and MAPK pathways.


Subject(s)
Cardiotonic Agents/pharmacology , Enkephalin, Methionine/pharmacology , ErbB Receptors/metabolism , Myocardial Ischemia/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , In Vitro Techniques , Ischemic Preconditioning, Myocardial , MAP Kinase Kinase Kinases/metabolism , Myocardial Ischemia/metabolism , Protein Kinase C/metabolism , Rabbits , Signal Transduction/drug effects
8.
Am J Physiol Heart Circ Physiol ; 288(4): H1717-23, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15550529

ABSTRACT

Recently, we reported that exogenous administration of Met(5)-enkephalin (ME) for 24 h reduces infarct size after ischemia-reperfusion in rabbits. In the present study, we tested whether ME-induced cardioprotection is exhibited in murine hearts and whether chronic infusion of this peptide can render hearts tolerant to ischemia. Barbiturate-anesthetized open-chest mice (C57BL/6J) were subjected to regional myocardial ischemia-reperfusion (45 min of occlusion and 20 min of reperfusion). Mice received saline vehicle or ME for 24 h or 2 wk before undergoing regional myocardial ischemia-reperfusion or for 24 h followed by a 24-h delay before regional myocardial ischemia-reperfusion. Infarct size was measured with propidium iodide and is expressed as a percentage of the area at risk. Infarcts were smaller after infusion of ME for 24 h than with vehicle control: 49.2 +/- 9.0% vs. 22.2 +/- 3.2% (P < 0.01). In contrast, administration of ME for 2 wk failed to elicit cardioprotection: 36.5 +/- 9.1% and 41.4 +/- 8.2% for control and ME, respectively (P = not significant). When a 24-h delay was imposed between the end of drug treatment and the onset of the ischemic insult, cardioprotection was lost: 38.5 +/- 6.1% and 42.8 +/- 6.6% for control and ME, respectively (P = not significant). Chronic sustained exogenous infusion of the endogenously produced opioid peptide ME is associated with loss of the cardioprotection that is observed with 24 h of infusion. Furthermore, in this in vivo murine model, ME failed to induce delayed tolerance to myocardial ischemia-reperfusion.


Subject(s)
Cardiotonic Agents/pharmacology , Enkephalin, Methionine/pharmacology , Myocardial Reperfusion Injury/drug therapy , Animals , Blood Pressure , Blotting, Western , Cyclic AMP/metabolism , Heart Rate , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Tachyphylaxis , Treatment Failure
9.
Coron Artery Dis ; 15(7): 449-55, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15492595

ABSTRACT

OBJECTIVES: We investigated the influence of the narcotic anesthetic remifentanil on irreversible myocardial ischemic injury. METHODS: New Zealand White rabbits were anesthetized with propofol (0.7-1.8 mg.kg.min) and then subjected to 30 min regional myocardial ischemia and 3 h reperfusion (CON). Some animals also underwent ischemic preconditioning, elicited by either one (IP1) or two (IP2) cycles of 5 min ischemia and 5 min reperfusion, and/or remifentanil, administered either as a transient infusion mimicking the preconditioning protocol (RP2, 10 microg x kg x min) or as a continuous infusion (R, 3-10 microg x kg x min). Rabbits were randomly assigned to experimental groups. Infarct size was assessed with tetrazolium. Results are reported as mean+/-SD. RESULTS: Non-preconditioned infarct size was approximately 50% of the area-at-risk (49.6+/-20.1% CON). Both one and two cycles of ischemic preconditioning markedly reduced infarct size (49.6+/-20.1% CON versus 18.6+/-8.6% IP and versus 7.5+/-7.6% IP2; both p<0.001). Preconditioning with remifentanil modestly reduced infarct size (49.6+/-20.1% CON versus 29.3+/-8.5% RP2; p<0.01). However, sustained administration of remifentanil did not provide protection (49.6+/-20.1% CON versus 43.9+/-16.2% R), and it attenuated the protection offered by preconditioning (49.6+/-20.1% CON versus 35.6+/-20.7% R+IP1, p=NS; and versus 14.5+/-14.5% R+IP2; p<0.05). CONCLUSION: Transient pre-ischemic administration of remifentanil modestly reduces infarct size in propofol-anesthetized rabbits, but continuous administration of remifentanil increases the threshold for ischemic preconditioning-induced infarct limitation.


Subject(s)
Anesthetics/therapeutic use , Ischemic Preconditioning, Myocardial , Myocardial Infarction/drug therapy , Piperidines/therapeutic use , Animals , Myocardial Infarction/pathology , Rabbits , Remifentanil
10.
Am J Physiol Heart Circ Physiol ; 286(6): H2229-36, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14726300

ABSTRACT

Regional changes occur in the sympathetic innervation of the heart after myocardial infarction (MI), including loss of norepinephrine (NE) uptake and depletion of neuronal NE. This apparent denervation is accompanied by increased cardiac NE spillover. One potential explanation for these apparently contradictory findings is that the sympathetic neurons innervating the heart are exposed to environmental stimuli that alter neuronal function. To understand the changes that occur in the innervation of the heart after MI, immunohistochemical, biochemical, and molecular analyses were carried out in the heart and stellate ganglia of control and MI rats. Immunohistochemistry with panneuronal markers revealed extensive denervation in the left ventricle (LV) below the infarct, but sympathetic nerve fibers were retained in the base of the heart. Western blot analysis revealed that tyrosine hydroxylase (TH) expression (normalized to a panneuronal marker) was increased significantly in the base of the heart and in the stellate ganglia but decreased in the LV below the MI. NE transporter (NET) binding sites, normalized to total protein, were unchanged, except in the LV, where [3H]nisoxetine binding was decreased. TH mRNA was increased significantly in the left and right stellate ganglia after MI, while NET mRNA was not. In the base of the heart, increased TH coupled with no change in NET may explain the increase in extracellular NE observed after MI. Coupled with substantial denervation in the LV, these changes likely contribute to the onset of cardiac arrhythmias.


Subject(s)
Adrenergic Fibers/pathology , Heart/innervation , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Norepinephrine/physiology , Adrenergic Fibers/physiology , Animals , Immunohistochemistry , Male , Norepinephrine Plasma Membrane Transport Proteins , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Symporters/genetics , Symporters/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Ubiquitin Thiolesterase/metabolism
11.
Am J Physiol Heart Circ Physiol ; 285(6): H2463-70, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12869377

ABSTRACT

The opioid antagonist naloxone abolishes infarct limitation by myocardial ischemic preconditioning, suggesting that one or more endogenous opioid peptides can mediate cardiac protection against ischemic damage. We tested the hypothesis that the naturally occurring opioid peptide Met5-enkephalin (ME) modulates myocardial infarct size in vivo. Experiments were conducted in barbiturate-anesthetized open-chest rabbits subjected to regional myocardial ischemia-reperfusion. ME was administered via osmotic minipump for 24 h. Infarct size was assessed with tetrazolium and is expressed as a percentage of the area at risk. Exogenous ME reduced the amount of the risk zone infarcted by approximately 60% compared with saline-treated controls. ME-induced protection was sensitive to opioid receptor blockade with naloxone [NAL 50 +/- 2% vs. ME + NAL 39 +/- 3%, P = not significant (NS)] and also to blockade of sarcolemmal and mitochondrial ATP-sensitive K+ (KATP) channels [5-hydroxydecanoate (5-HD) 33 +/- 3% vs. ME + 5-HD 43 +/- 8%, P = NS; and HMR-1098 60 +/- 3% vs. ME + HMR-1098 54 +/- 7%, P = NS]. We conclude that ME limits ischemic injury in vivo by an opioid receptor-mediated mechanism that involves both sarcolemmal and mitochondrial KATP channels.


Subject(s)
Cardiotonic Agents/pharmacology , Enkephalin, Methionine/pharmacology , Ischemic Preconditioning , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/drug therapy , Animals , Body Temperature , Coronary Disease/drug therapy , Coronary Disease/pathology , Heart Rate , Male , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Potassium Channels/metabolism , Rabbits
12.
Am J Physiol Heart Circ Physiol ; 285(3): H1032-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12730057

ABSTRACT

To examine the receptor specificity and the mechanism of opioid peptide-induced protection, we examined freshly isolated adult rabbit cardiomyocytes subjected to simulated ischemia. Cell death as a function of time was assessed by trypan blue permeability. Dynorphin B (DynB) and Met5-enkephalin (ME) limitation of cell death (expressed as area under the curve) was sensitive to blockade by naltrindole (NTI, a delta-selective antagonist) and 5'-guanidinyl-17-(cyclopropylmethyl)-6,7-dehydro-4,5alpha-epoxy-3,14-dihydroxy-6,7-2',3'-indolomorphinan (GNTI dihydrochloride, a kappa-selective antagonist): 85.7 +/- 2.7 and 142.9 +/- 2.7 with DynB and DynB + NTI, respectively (P < 0.001), 94.1 +/- 4.2 and 164.5 +/- 7.3 with DynB and DynB + GNTI, respectively (P < 0.001), 111.9 +/- 7.0 and 192.1 +/- 6.4 with ME and ME + NTI, respectively (P < 0.001), and 120.2 +/- 4.3 and 170.0 +/- 3.3 with ME and ME + GNTI, respectively (P < 0.001). Blockade of ATP-sensitive K+ channels eliminated DynB- and ME-induced protection: 189.6 +/- 5.4 and 139.0 +/- 5.4 for control and ME, respectively (P < 0.001), and 210 +/- 5.9 and 195 +/- 6.1 for 5-HD and ME + 5-HD, respectively (P < 0.001); 136.0 +/- 5.7 and 63.4 +/- 5.4 for control and ME, respectively (P < 0.001), and 144.6 +/- 4.5 and 114.6 +/- 7.7 for HMR-1098 and ME + HMR-1098, respectively (P < 0.01); 189.6 +/- 5.4 and 139.0 +/- 5.4 for control and ME, respectively (P < 0.001), and 210 +/- 5.9 and 195 +/- 6.1 for 5-HD and ME + 5-HD, respectively (P < 0.001); and 136.0 +/- 5.7 and 63.4 +/- 5.4 for control and ME, respectively (P < 0.001), and 144.6 +/- 4.5 and 114.6 +/- 7.7 for HMR-1098 and ME + HMR-1098, respectively (P < 0.01). We conclude that opioid peptide-induced cardioprotection is mediated by delta- and kappa-receptors and involves sarcolemmal and mitochondrial ATP-sensitive K+ channels.


Subject(s)
Dynorphins/pharmacology , Endorphins/pharmacology , Enkephalin, Methionine/pharmacology , Myocardial Ischemia/metabolism , Naltrexone/analogs & derivatives , Potassium Channels/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Adenosine Triphosphate/metabolism , Animals , Benzamides/pharmacology , Cardiotonic Agents/pharmacology , Dose-Response Relationship, Drug , Guanidines , Ischemic Preconditioning, Myocardial , Male , Mitochondria/metabolism , Morphinans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Rabbits , Sarcolemma/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...