Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Thromb Haemost ; 112(6): 1110-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25354936

ABSTRACT

Despite optimal therapy, the morbidity and mortality of patients presenting with an acute myocardial infarction (MI) remain significant, and the initial mechanistic trigger of myocardial "ischaemia/reperfusion (I/R) injury" remains greatly unexplained. Here we show that factors released from the damaged cardiac tissue itself, in particular extracellular RNA (eRNA) and tumour-necrosis-factor α (TNF-α), may dictate I/R injury. In an experimental in vivo mouse model of myocardial I/R as well as in the isolated I/R Langendorff-perfused rat heart, cardiomyocyte death was induced by eRNA and TNF-α. Moreover, TNF-α promoted further eRNA release especially under hypoxia, feeding a vicious cell damaging cycle during I/R with the massive production of oxygen radicals, mitochondrial obstruction, decrease in antioxidant enzymes and decline of cardiomyocyte functions. The administration of RNase1 significantly decreased myocardial infarction in both experimental models. This regimen allowed the reduction in cytokine release, normalisation of antioxidant enzymes as well as preservation of cardiac tissue. Thus, RNase1 administration provides a novel therapeutic regimen to interfere with the adverse eRNA-TNF-α interplay and significantly reduces or prevents the pathological outcome of ischaemic heart disease.


Subject(s)
Autocrine Communication/drug effects , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , Myocytes, Cardiac/drug effects , RNA/metabolism , Ribonucleases/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Antioxidants/metabolism , Cell Hypoxia , Cytoprotection , Disease Models, Animal , Mice , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Mitochondria, Heart/pathology , Myocardial Infarction/enzymology , Myocardial Infarction/genetics , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Myocardium/immunology , Myocardium/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , RNA/genetics , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Time Factors , Tumor Necrosis Factor-alpha/immunology
2.
Cardiovasc Res ; 51(3): 592-600, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11476750

ABSTRACT

OBJECTIVE: It has been shown that cGMP content is reduced in post-ischemic myocardium, and that stimulation of cGMP synthesis prevents cardiomyocyte hypercontracture and cell death in vitro. This study was aimed at determining whether administration of the natriuretic peptide urodilatin (URO) at the time of reperfusion could limit myocardial cell death secondary to transient coronary occlusion. METHODS: The relation between cGMP content in reperfused myocardium and the extent of cell death was investigated in isolated rat hearts (n=62) receiving different URO concentrations during initial reperfusion. The dose of intravenous URO necessary to obtain the targeted increase in cGMP in reperfused myocardium was investigated in ten pigs submitted to transient coronary occlusion (CO), and the effect of two selected doses of URO on infarct size was investigated in 22 pigs. RESULTS: cGMP was severely reduced in post-ischemic rat hearts. Addition of 0.01 microM URO during the first 15 min of reperfusion had no effect on myocardial cGMP content, functional recovery or LDH release in hearts submitted to 40 or 60 min of ischemia. At 0.05 microM, URO increased myocardial cGMP to 111% of values in normoxic hearts, improved functional recovery (P=0.01) and reduced peak LDH released by 40% (P=0.02). The beneficial effect of urodilatin was abolished by ANP receptor inhibition. At 1 microM, URO increased cGMP in reperfused myocardium to 363% of normoxic controls and had no beneficial effect. In pigs allocated to 47 min of CO and 5 min of reperfusion, cGMP was markedly reduced in reperfused myocardium. Intravenous URO at 10 ng/kg per min during the first 25 min of reperfusion normalized myocardial cGMP after 5 min of reflow (95% of control myocardium), and reduced infarct size by 40% (P=0.04). At 50 ng/kg per min, urodilatin increased myocardial cGMP in reperfused myocardium to 335% of control myocardium and failed to significantly reduce infarct size (46 vs. 66%, P=0.125). None of these doses had detectable hemodynamic effects. CONCLUSIONS: Intravenous low-dose URO at the time of reperfusion normalizes myocardial cGMP and limits necrosis. Large doses of URO increasing myocardial cGMP well over normal values may lack this beneficial effect.


Subject(s)
Atrial Natriuretic Factor/therapeutic use , Diuretics/therapeutic use , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Peptide Fragments/therapeutic use , Animals , Atrial Natriuretic Factor/administration & dosage , Atrial Natriuretic Factor/blood , Coronary Circulation/drug effects , Cyclic GMP/metabolism , Diuretics/administration & dosage , Diuretics/blood , Dose-Response Relationship, Drug , Hemodynamics/drug effects , Infusions, Intravenous , L-Lactate Dehydrogenase/metabolism , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Organ Culture Techniques , Peptide Fragments/administration & dosage , Peptide Fragments/blood , Swine
3.
Cardiovasc Res ; 51(2): 241-50, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11470463

ABSTRACT

OBJECTIVE: We have recently shown that spontaneous Ca2+ oscillations elicit irreversible hypercontracture of cardiomyocytes during reoxygenation. The aim of this study was to investigate whether influx of exterior Ca2+ through the reverse mode of the Na+/Ca2+ exchanger (NCE) contributes to the development of these oscillations and, therefore, to reoxygenation-induced hypercontracture. METHODS: Isolated cardiomyocytes and hearts from rats were used as models. Cardiomyocytes were exposed to 60 min simulated ischemia (pH(o) 6.4) and 10 min reoxygenation (pH(o) 7.4). During reoxygenation cardiomyocytes were superfused with medium containing 1 mmol/l Ca2+ (control), with nominally Ca2+-free medium or with medium containing 10 micromol/l KB-R 7943 (KB), a selective inhibitor of the reverse mode of the NCE. RESULTS: In reoxygenated cardiomyocytes rapid Ca2+ oscillations occurred which were reduced under Ca2+-free conditions or in presence of KB. Hypercontracture was also significantly reduced under Ca2+-free conditions or in presence of KB. After 30 min of normoxic perfusion isolated rat hearts were subjected to 60 min global ischemia and reperfusion. KB (10 micromol/l) was present during the first 10 min of reperfusion. LVEDP, LVdevP and lactate dehydrogenase (LDH) release were measured. Presence of KB reduced post-ischemic LVEDP and improved left ventricular function (LVdevP). In KB treated hearts the reperfusion induced release of LDH was markedly reduced from 81.1 +/- 9.9 (control) to 49.3 +/- 8.8 U/60 min/g dry weight. CONCLUSION: Our study shows that inhibition of the reverse mode of the NCE, during reperfusion only, protects cardiomyocytes and whole hearts against reperfusion injury.


Subject(s)
Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Sodium-Calcium Exchanger/antagonists & inhibitors , Thiourea/pharmacology , Analysis of Variance , Animals , Calcium/metabolism , Cell Size , Cells, Cultured , Cytosol/metabolism , Hydrogen-Ion Concentration , Male , Models, Animal , Myocardial Reperfusion Injury/prevention & control , Perfusion , Rats , Rats, Sprague-Dawley , Rats, Wistar , Sodium/metabolism , Thiourea/analogs & derivatives , Time Factors
4.
Am J Physiol Heart Circ Physiol ; 280(6): H2563-71, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356611

ABSTRACT

During myocardial ischemia, severe ATP depletion induces rigor contracture followed by intracellular Ca2+ concentration ([Ca2+]i) rise and progressive impairment of gap junction (GJ)-mediated electrical coupling. Our objective was to investigate whether chemical coupling through GJ allows propagation of rigor in cardiomyocytes and whether it persists after rigor development. In end-to-end connected adult rat cardiomyocytes submitted to simulated ischemia the interval between rigor onset was 3.7 +/- 0.7 s, and subsequent [Ca2+]i rise was virtually identical in both cells, whereas in nonconnected cell pairs the interval was 71 +/- 12 s and the rate of [Ca2+]i rise was highly variable. The GJ blocker 18alpha-glycyrrhetinic acid increased the interval between rigor onset and the differences in [Ca(2+)]i between connected cells. Transfer of Lucifer yellow demonstrated GJ permeability 10 min after rigor onset in connected cell pairs, and 30 min after rigor onset in isolated rat hearts submitted to nonflow ischemia but was abolished after 2 h of ischemia. GJ-mediated communication allows propagation of rigor in ischemic myocytes and persists after rigor development despite acidosis and increased [Ca2+]i.


Subject(s)
Cell Communication , Gap Junctions/metabolism , Myocardial Ischemia/metabolism , Animals , Calcium/metabolism , Cell Membrane Permeability , Cells, Cultured , Fluorescent Dyes , Gap Junctions/drug effects , Glycyrrhetinic Acid/analogs & derivatives , Glycyrrhetinic Acid/pharmacology , Heart Ventricles/cytology , Heart Ventricles/metabolism , In Vitro Techniques , Male , Myocardial Contraction , Rats , Rats, Sprague-Dawley , Time Factors
5.
Cardiovasc Res ; 46(3): 412-20, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10912452

ABSTRACT

OBJECTIVE: Stimulation of cGMP synthesis protects cardiomyocytes against reoxygenation-induced hypercontracture. The purpose of this study was to determine whether L-arginine supplementation has a protective effect against reperfusion-induced hypercontracture and necrosis in the intact animal. METHODS: Twenty-four Large-White pigs were randomized to receive either 100 mg/kg of L-arginine i.v. or vehicle 10 min before 48 min of coronary occlusion and 2 h of reperfusion. Hemodynamic variables, coronary blood flow and myocardial segment length changes (piezoelectric crystals) were monitored. Postmortem studies included quantification of myocardium at risk (in vivo fluorescein), infarct size (triphenyltetrazolium reaction), myocardial myeloperoxidase activity and histological analysis. Systemic, coronary vein, and myocardial cGMP concentration were measured in additional animals. RESULTS: Administration of L-arginine had no significant effect in hemodynamics or coronary blood flow. During reperfusion, myocardial cGMP content was reduced in the LAD as compared to control myocardium (P=0.02). L-Arginine increased myocardial cGMP content and caused a transient increase in plasma cGMP concentration during the initial minutes of reperfusion (P=0.02). The reduction in end-diastolic segment length induced by reperfusion, reflecting hypercontracture, was less pronounced in the L-arginine group (P=0.02). Infarct size was smaller in pigs receiving L-arginine (47.9+/-7.2% of the area at risk) than in controls (62.9+/-4.9%, P=0.047). There were no differences between groups in leukocyte accumulation in reperfused myocardium (P=0.80). CONCLUSION: L-Arginine supplementation reduces myocardial necrosis secondary to in situ ischemia-reperfusion by a direct protective effect against myocyte hypercontracture.


Subject(s)
Arginine/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/prevention & control , Analysis of Variance , Animals , Arginine/blood , Blood Pressure/drug effects , Cell Size/drug effects , Cyclic GMP/blood , Female , Heart Rate/drug effects , Male , Myocardial Contraction/drug effects , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Peroxidase/metabolism , Random Allocation , Swine
6.
Cardiovasc Res ; 45(2): 351-9, 2000 Jan 14.
Article in English | MEDLINE | ID: mdl-10728355

ABSTRACT

OBJECTIVES: Hypercontracture is an important mechanism of myocyte death during reperfusion. cGMP modulates the sensitivity of contractile myofilaments to Ca2+, and increasing cGMP concentration during the last minutes of anoxia prevents reoxygenation-induced hypercontracture in isolated cardiomyocytes. The purpose of this study was to determine whether stimulation of particulate guanylyl cyclase with the natriuretic peptide urodilatin, given at the time of reperfusion, reduces myocardial necrosis in the rat heart submitted to transient ischemia. METHODS: Isolated rat hearts (n = 38) were submitted to either 40 or 60 min of no-flow ischemia and 2 h of reperfusion, and were allocated to receive or not receive 0.05 microM urodilatin during the first 15 min of reperfusion or non-reperfusion treatment. RESULTS: A marked reduction in myocardial cGMP concentration was observed in control hearts during reperfusion after 40 or 60 min of ischemia. Urodilatin significantly attenuated cGMP depletion during initial reperfusion, markedly improved contractile recovery after 40 min of ischemia (P < 0.0309), and reduced reperfusion-induced increase in left ventricular end-diastolic pressure (P = 0.0139), LDH release (P = 0.0263), and contraction band necrosis (P = 0.0179) after 60 min of ischemia. The beneficial effect of urodilatin was reproduced by the membrane permeable cGMP analog 8-Bromo-cGMP. CONCLUSIONS: These results indicate that reduced cGMP concentration may impair myocyte survival during reperfusion. Stimulation of particulate guanylyl cyclase may appear as a new strategy to prevent immediate lethal reperfusion injury.


Subject(s)
Atrial Natriuretic Factor/therapeutic use , Enzyme Activators/therapeutic use , Guanylate Cyclase/metabolism , Myocardial Reperfusion Injury/prevention & control , Peptide Fragments/therapeutic use , Analysis of Variance , Animals , Cyclic GMP/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Necrosis , Perfusion , Rats , Rats, Sprague-Dawley
7.
Exp Physiol ; 85(1): 17-25, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10662888

ABSTRACT

The objective of this study was to investigate the effect of Na+-H+ exchange (NHE) and HCO3--Na+ symport inhibition on the development of rigor contracture. Freshly isolated adult rat cardiomyocytes were subjected to 60 min metabolic inhibition (MI) and 5 min re-energization (Rx). The effects of perfusion of HCO3- or HCO3--free buffer with or without the NHE inhibitor HOE642 (7 microM) were investigated during MI and Rx. In HCO3--free conditions, HOE642 reduced the percentage of cells developing rigor during MI from 79 +/- 1% to 40 +/- 4% (P < 0.001) without modifying the time at which rigor appeared. This resulted in a 30% reduction of hypercontracture during Rx (P < 0.01). The presence of HCO3- abolished the protective effect of HOE642 against rigor. Cells that had developed rigor underwent hypercontracture during Rx independently of treatment allocation. Ratiofluorescence measurement demonstrated that the rise in cytosolic Ca2+ (fura-2) occurred only after the onset of rigor, and was not influenced by HOE642. NHE inhibition did not modify Na+ rise (SBFI) during MI, but exaggerated the initial fall of intracellular pH (BCEFC). In conclusion, HOE642 has a protective effect against rigor during energy deprivation, but only when HCO3--dependent transporters are inhibited. This effect is independent of changes in cytosolic Na+ or Ca2+ concentrations.


Subject(s)
Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Heart/drug effects , Myocardial Contraction/drug effects , Myocardium/cytology , Rigor Mortis/prevention & control , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sulfones/pharmacology , Animals , Bicarbonates/metabolism , Calcium/metabolism , Energy Metabolism/physiology , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/enzymology , Hydrogen-Ion Concentration , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Male , Myocardium/enzymology , Perfusion , Rats , Rats, Sprague-Dawley , Rigor Mortis/enzymology , Sodium/metabolism
8.
Am J Physiol Heart Circ Physiol ; 278(1): H94-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10644588

ABSTRACT

Oxidative stress may cause apoptosis of cardiomyocytes in ischemic-reperfused myocardium. We investigated whether ischemia-reperfusion modifies the susceptibility of cardiomyocyte induction of apoptosis by oxidative stress. Ischemia was simulated by incubating isolated cardiomyocytes from adult rats in an anoxic, glucose-free medium, pH 6.4, for 3 h. Annexin V-fluorescein isothiocyanate/propidium iodide staining and the detection of DNA laddering were used as apoptotic markers. H(2)O(2) (7.5 micromol/l) induced apoptosis in 20.1 +/- 1.8% of cells under normoxic conditions but only 14.4 +/- 1.6% (n = 6, P < 0.05) after ischemia-reoxygenation. This partial protection of ischemic-reoxygenated cells was observed despite a reduction in their cellular glutathione content, from 11.4 +/- 1.9 in normoxic controls to 2.9 +/- 0.8 nmol/mg protein (n = 3, P < 0.05). Elevation of end-ischemic glutathione contents by pretreatment with 1 mmol/l N-acetylcysteine entirely protected ischemic-reoxygenated cells against induction of apoptosis by H(2)O(2). In conclusion, ischemia-reperfusion can protect cardiomyocytes against induction of apoptosis by exogenous oxidative stress. This endogenous protective effect is most clearly demonstrated when control and postischemic cardiomyocytes are compared at similar glutathione levels.


Subject(s)
Apoptosis , Myocardial Ischemia/physiopathology , Oxidative Stress , Acetylcysteine/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Free Radical Scavengers/pharmacology , Hydrogen Peroxide/pharmacology , Male , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Oxidants/pharmacology , Phosphocreatine/pharmacology , Rats , Rats, Wistar
9.
J Exp Med ; 189(9): 1373-82, 1999 May 03.
Article in English | MEDLINE | ID: mdl-10224277

ABSTRACT

The effect of factor XIII on endothelial barrier function was studied in a model of cultured monolayers of porcine aortic endothelial cells and saline-perfused rat hearts. The thrombin-activated plasma factor XIII (1 U/ml) reduced albumin permeability of endothelial monolayers within 20 min by 30 +/- 7% (basal value of 5.9 +/- 0.4 x 10(-6) cm/s), whereas the nonactivated plasma factor XIII had no effect. Reduction of permeability to the same extent, i.e., by 34 +/- 9% could be obtained with the thrombin-activated A subunit of factor XIII (1 U/ml), whereas the iodoacetamide-inactivated A subunit as well as the B subunit had no effect on permeability. Endothelial monolayers exposed to the activated factor XIII A exhibited immunoreactive deposition of itself at interfaces of adjacent cells; however, these were not found on exposure to nonactivated factor XIII A or factor XIII B. Hyperpermeability induced by metabolic inhibition (1 mM potassium cyanide plus 1 mM 2-deoxy-D-glucose) was prevented in the presence of the activated factor XIII A. Likewise, the increase in myocardial water content in ischemic-reperfused rat hearts was prevented in its presence. This study shows that activated factor XIII reduces endothelial permeability. It can prevent the loss of endothelial barrier function under conditions of energy depletion. Its effect seems related to a modification of the paracellular passageways in endothelial monolayers.


Subject(s)
Endothelium, Vascular/physiology , Factor XIII/metabolism , Animals , Aorta/cytology , Body Water , Cell Membrane Permeability , Cells, Cultured , Endothelium, Vascular/cytology , Male , Rats , Rats, Wistar , Staining and Labeling , Swine
10.
Am J Physiol ; 276(5): H1574-80, 1999 05.
Article in English | MEDLINE | ID: mdl-10330241

ABSTRACT

The objective of this study was to investigate the effect of L-arginine supplementation on myocardial cell death secondary to hypoxia-reoxygenation. Isolated rat hearts (n = 51) subjected to 40 min of hypoxia and 90 min of reoxygenation received 3 mM L-arginine and/or 1 microM 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ; a selective inhibitor of soluble guanylyl cyclase) throughout the experiment or during the equilibration, hypoxia, or reoxygenation periods. The incorporation of L-[3H]arginine into myocytes during energy deprivation was investigated in isolated adult rat myocytes. The addition of L-arginine to the perfusate throughout the experiment resulted in higher cGMP release (P < 0.05), reduced lactate dehydrogenase release (P < 0.05), and increased pressure-rate product (P < 0.05) during reoxygenation. These effects were reproduced when L-arginine was added only during equilibration, but addition of L-arginine during hypoxia or reoxygenation had no effect. Addition of ODQ either throughout the experiment or only during reoxygenation reversed the beneficial effects of L-arginine. L-[3H]arginine was not significantly incorporated into isolated myocytes subjected to energy deprivation. We conclude that L-arginine supplementation protects the myocardium against reoxygenation injury by cGMP-mediated actions. To be effective during reoxygenation, L-arginine must be added before anoxia.


Subject(s)
Arginine/pharmacokinetics , Cyclic GMP/metabolism , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Animals , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Hypoxia/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Muscle Fibers, Skeletal/enzymology , Myocardial Contraction/physiology , Myocardium/cytology , Nitric Oxide/metabolism , Oxadiazoles/pharmacology , Oxygen Consumption/physiology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley
11.
Biochem Pharmacol ; 57(1): 111-20, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9920291

ABSTRACT

Tetanus toxin (TeTx) is a powerful clostridial neurotoxin that inhibits Ca2+-dependent neurotransmitter secretion as do the botulinum neurotoxins (BoNTs). We found that TeTx (but not BoNT/A) produced a specific time- and dose-dependent inhibition of Na+-dependent [3H]5-hydroxytryptamine (serotonin, 5-HT) uptake in rat CNS synaptosomes. This effect was found in all CNS tryptaminergic areas, being maximal in the hippocampus and occipital cortex. TeTx produced the maximum reduction in [3H]5-HT uptake after 30 min of preincubation, being significant also at lower doses (10(-12) M) or shorter incubation times (10 min). Serotonin transport inhibitors such as fenfluramine (IC50, 11.0 +/- 0.9 microM), paroxetine (IC50, 33.5 +/- 0.1 microM), and imipramine (IC50, 89.9 +/- 5.7 microM) were 3 or 4 orders of magnitude less potent than TeTx (IC50, 8.7 +/- 1.0 nM). Of the two fragments of TeTx, (the C-terminal portion of the neurotoxin heavy chain, which is responsible for the binding to the nerve tissue) was consistently more effective than the L-H(N) fragment (the light neurotoxin chain disulfide linked to the N-terminal portion of the heavy chain, which is responsible for the toxic metalloprotease action) as inhibitor of [3H]5-HT uptake in synaptosomal preparations (56 +/- 5% and 95 +/- 3% with respect to control, respectively). Antagonism of the toxin-induced [3H]5-HT uptake blockade could not be reversed by zinc chelators but did have the ability to antagonize the TeTx inhibition of basal and K+-evoked [3H]5-HT release in rat synaptosomes. The reduction in serotonin accumulation induced by TeTx could be responsible for some tetanic symptoms that have been related to the serotonergic system.


Subject(s)
Brain/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin/metabolism , Sodium/pharmacology , Synaptosomes/metabolism , Tetanus Toxin/pharmacology , Animals , Biological Transport/drug effects , Botulinum Toxins/pharmacology , Captopril/pharmacology , Fenfluramine/pharmacology , Imipramine/pharmacology , Kinetics , Male , Organ Specificity , Paroxetine/pharmacology , Phenanthrolines/pharmacology , Rats , Rats, Sprague-Dawley , Synaptosomes/drug effects , Temperature , Tritium
12.
J Thorac Cardiovasc Surg ; 115(3): 709-15, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9535460

ABSTRACT

OBJECTIVE: The hypothesis was that Na+/H+ exchange occurring during normothermic cardioplegia contributes to the development of myocardial edema during subsequent reperfusion and impairs functional recovery. METHODS: Rat hearts were perfused in a Langendorff apparatus and submitted to 60 minutes of normothermic cardioplegia and 90 minutes of reperfusion. Hearts were allocated to one of four groups (n = 8): inhibition of Na+/H+ exchanger with HOE642 throughout the whole experiment (HOE group), only during cardioplegia (HOE-C) or during reperfusion (HOE-R), and a control group. RESULTS: In HOE and HOE-C groups, myocardial water content at the end of reperfusion was lower than in the HOE-R and control groups (526 +/- 19 and 533 +/- 18 ml/100 gm dry tissue vs 632 +/- 25 and 634 +/- 17 ml/100 gm dry tissue, respectively, p = 0.001), left ventricular end-diastolic pressure increased less after reperfusion (46.6 +/- 9.7 and 63.2 +/- 10.0 mm Hg vs 75.1 +/- 4.3 mm Hg and 85.7 +/- 8.9 mm Hg, respectively, p = 0.006), and recovery of left ventricular developed pressure was better (46.7% and 45.8% vs 4.5% and 9.8%, p = 0.048). Relative to the control group, total lactate dehydrogenase release during reperfusion was reduced by 80.2%, 69.3% and 36% in HOE, HOE-C, and HOE-R groups, respectively. CONCLUSION: Inhibition of the Na+/H+ exchange during normothermic cardioplegia reduces myocardial edema and necrosis during subsequent reperfusion, improving functional recovery. Inhibition of Na+/H+ exchange during reperfusion only has a much smaller effect.


Subject(s)
Edema, Cardiac/prevention & control , Guanidines/pharmacology , Heart Arrest, Induced , Myocardial Reperfusion Injury/prevention & control , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sulfones/pharmacology , Animals , Edema, Cardiac/etiology , Edema, Cardiac/physiopathology , Hemodynamics , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/physiopathology , Rats , Rats, Sprague-Dawley , Ventricular Function, Left
13.
Circulation ; 96(10): 3579-86, 1997 Nov 18.
Article in English | MEDLINE | ID: mdl-9396458

ABSTRACT

BACKGROUND: The objective of this study was to test the hypothesis that chemical interaction through gap junctions may result in cell-to-cell progression of hypercontracture and that this phenomenon contributes to the final extent of reperfused infarcts. METHODS AND RESULTS: Cell-to-cell transmission of hypercontracture was studied in pairs of freshly isolated adult rat cardiomyocytes. Hypercontracture induced by microinjection of a solution containing 1 mmol/L Ca2+ and 2% lucifer yellow (LY) was transmitted to the adjacent cell (11 of 11 pairs), and the gap junction uncoupler heptanol (2 mmol/L) prevented transmission in 6 of 8 pairs (P=.003), with a perfect association between passage of the LY and transmission of hypercontracture. In the isolated, perfused rat heart submitted to 30 minutes of hypoxia, addition of heptanol to the perfusion media during the first 15 minutes of reoxygenation had a dose-related protective effect against the oxygen paradox, as demonstrated by a reduction of diastolic pressure and marked recovery of developed pressure (P<.001), as well as less lactate dehydrogenase release during reoxygenation (P<.001) and less contraction band necrosis (P<.001) than controls. In the in situ pig heart submitted to 48 minutes of coronary occlusion, the intracoronary infusion of heptanol during the first 15 minutes of reperfusion at a final concentration of 1 mmol/L limited myocardial shrinkage, reflecting hypercontracture (P<.05), reduced infarct size after 5 hours of reperfusion by 54% (P=.04), and modified infarct geometry with a characteristic fragmentation of the area of necrosis. Heptanol at 1 mmol/L had no significant effect on contractility of nonischemic myocardium. CONCLUSIONS: These results demonstrate that hypercontracture may be transmitted to adjacent myocytes through gap junctions and that heptanol may interfere with this transmission and reduce the final extent of myocardial necrosis during reoxygenation or reperfusion. These findings are consistent with the hypothesis tested and open a new approach to limitation of infarct size by pharmacological control of gap junction conductance.


Subject(s)
Gap Junctions/drug effects , Heptanol/pharmacology , Myocardial Contraction/drug effects , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Uncoupling Agents/pharmacology , Animals , Cell Separation , Coronary Circulation/drug effects , Coronary Disease/physiopathology , Disease Progression , Heart/drug effects , Hemodynamics/drug effects , Hypoxia/physiopathology , In Vitro Techniques , Necrosis , Rats , Rats, Sprague-Dawley
14.
J Mol Cell Cardiol ; 29(4): 1167-75, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9160868

ABSTRACT

To investigate the role of Na(+)-H+ exchange occurring during hypoxia in the genesis of reoxygenation-induced myocardial oedema, isolated perfused rat hearts were submitted to 40 min of hypoxia and 90 min of reoxygenation. The influence of three factors on myocardial water content was analysed according to a 2 x 2 x 2 factorial design; the hearts were perfused at either pH = 7.4 or pH = 7.0, with either HCO3- buffer or HCO3(-)-free HEPES buffer, and in half of the experiments the hypoxic buffer contained HOE642 6.7 micromol/l. In an additional group, 160 min of normoxia resulted in no lactate dehydrogenase (LDH) release and in a 35.8% increase in myocardial water, independently of pH and of the presence of HCO3- in the buffer. In hearts perfused at pH = 7.4, reoxygenation induced LDH release which was reduced (P<0.05) by HOE642 by 20.1%, by HCO3(-)-free perfusion by 57.5%, and by the combination of both by 91.2%. Reoxygenation also induced severe myocardial oedema (26.3% increase (P<0.05) respect to normoxia). HOE642 reduced (P<0.05) reoxygenation oedema by 15.7%, HCO3(-)-free perfusion by 8.9%, and the combination of both by 24.6%. The effects of HCO3(-)-free perfusion could be mimicked in HCO3(-)-perfused hearts by blocking Na(+)-HCO3- cotransport with 4-4'-dibenzanidostilbene-2,2'-disulphonic acid (DIDS). The beneficial and additive effects of HOE642 and of HCO3(-)-free perfusion on oedema were not a mere consequence of their protective effects against the oxygen paradox, since they were observed in groups perfused at pH= 7.0, a condition which virtually prevented LDH release without preventing oedema (19.0% increase in myocardial water). Thus, reoxygenation-induced myocardial oedema may occur in the absence of necrosis, and is largely determined by Na+ gain during hypoxia via Na(+)-H+ exchange and Na(+)-HCO3- cotransport.


Subject(s)
Cardiomyopathies/metabolism , Edema/metabolism , Hypoxia/metabolism , Oxygen/physiology , Sodium-Hydrogen Exchangers/physiology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Antiporters/drug effects , Antiporters/metabolism , Bicarbonates/metabolism , Buffers , Cardiomyopathies/physiopathology , Edema/physiopathology , Guanidines/pharmacology , Hydrogen-Ion Concentration , Hypoxia/drug therapy , In Vitro Techniques , L-Lactate Dehydrogenase/drug effects , L-Lactate Dehydrogenase/metabolism , Male , Perfusion , Rats , Rats, Sprague-Dawley , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sulfones/pharmacology , Ventricular Function, Left , Ventricular Pressure , Water/chemistry
15.
Cardiovasc Res ; 32(3): 587-92, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8881519

ABSTRACT

OBJECTIVE: Cytoskeletal and sarcolemmal fragility secondary to anoxia may contribute to sarcolemmal rupture and cell death during reoxygenation of cardiomyocytes. This study investigated the influence of trimetazidine (TMZ), a drug with effects on lipid metabolism and cell membranes, on reoxygenation-induced sarcolemmal rupture. METHODS: Isolated adult rat myocytes were submitted to 60 min of metabolic inhibition and 5 min of hypo-osmotic reoxygenation to simulate reperfusion edema in situ. Cells were allocated to 3 groups of treatment: in one group, TMZ 100 mumol/l was added to both the metabolic inhibition and reoxygenation buffers (group TMZ); another group was submitted to the same treatment but cells had previously been incubated with TMZ 100 mumol/l for 3 h (group TMZ-Pre); a control group underwent metabolic inhibition and hypo-osmotic reoxygenation without any treatment. Cell morphology was monitored throughout the experiment and sarcolemmal integrity was assessed by quantification of LDH activity and trypan blue exclusion test. RESULTS: After 60 min of metabolic inhibition most cells (83.1 +/- 2%) presented rigor contracture without between-group differences. Reoxygenation resulted in hypercontracture of 84.2 +/- 2.3, 91.2 +/- 1.4 and 84.1 +/- 2.1% of cells in TMZ, TMZ-Pre and control groups, P = NS. The trypan blue exclusion test revealed a higher proportion of cells with sarcolemmal integrity in TMZ and TMZ-Pre groups than in controls (12.7 +/- 2.0, 10.0 +/- 1.5 and 6.3 +/- 0.8%, respectively, P = 0.002). No between-group differences in LDH activity in the extracellular medium were observed at the onset or at the end of metabolic inhibition. However, LDH release was significantly lower (P = 0.002) in the TMZ-Pre group (1.6 +/- 0.1 IU/1000 cells) than in the TMZ and control groups (1.9 +/- 0.2 and 2.2 +/- 0.1 IU/1000 cells). CONCLUSION: Preincubation of cardiomyocytes with TMZ does not prevent rigor contracture induced by metabolic inhibition or hypercontracture during subsequent reoxygenation, but does improve sarcolemmal resistance to reoxygenation-induced mechanical stress. This could help to explain the beneficial effect of TMZ on infarct size.


Subject(s)
Myocardial Ischemia/metabolism , Myocardium/metabolism , Oxygen/metabolism , Sarcolemma/metabolism , Trimetazidine/pharmacology , Animals , Cells, Cultured , Heart/drug effects , Male , Myocardial Reperfusion Injury/prevention & control , Osmotic Pressure , Rats , Rats, Sprague-Dawley , Sarcolemma/drug effects , Stress, Mechanical , Video Recording
SELECTION OF CITATIONS
SEARCH DETAIL
...