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










Publication year range
1.
J Physiol Pharmacol ; 66(2): 215-26, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25903952

ABSTRACT

During ischaemia/reperfusion, the rise in [Na(+)](i), induced by simultaneous depression of the Na(+)/K(+)-ATPase and activation of the Na(+)/H(+) exchanger (NHE), shifts the Na(+)/Ca(2+) exchanger (NCX) into reverse transport mode, resulting in Ca(2+)(i)overload, which is a critical factor in enhancing the liability to cardiac arrhythmias. The inhibition of NHE, and recently NCX has been suggested to effectively protect the heart from reperfusion-induced arrhythmias. In this study, we investigated and compared the efficacy of individual or the simultaneous inhibition of the NHE and NCX against reperfusion-induced arrhythmias in Langendorff-perfused rat hearts by applying a commonly used regional ischaemia-reperfusion protocol. The NHE and NCX were inhibited by cariporide and SEA0400 or the novel, more selective ORM-10103, respectively. Arrhythmia diagrams calculated for the reperfusion period were analysed for the incidence and duration of extrasystoles (ESs), ventricular tachycardia (VT) and ventricular fibrillation (VF). NHE inhibition by cariporide was highly efficient in reducing the recorded reperfusion-induced arrhythmias. Following the application of SEA0400 or ORM-10103, the number and duration of arrhythmic periods were efficiently or moderately decreased. While both NCX inhibitors effectively reduced ESs, the most frequently triggered arrhythmias, they exerted limited or no effect on VTs and VFs. Of the NCX inhibitors, ORM-10103 was more effective. Surprisingly, the simultaneous inhibition of the NCX and NHE failed to significantly improve the antiarrhythmic efficacy reached by NCX blockade alone. In conclusion, although principal simultaneous NHE+NCX inhibition should be highly effective against all types of the recorded reperfusion-induced arrhythmias, NCX inhibitors, alone or in combination with cariporide, seem to be moderately suitable to provide satisfactory cardioprotection - at least in the present arrhythmia model. Since ORM-10103 and SEA0400 are known to effectively inhibit after-depolarisations, it is suggested that their efficacy and that of other NCX inhibitors may be higher and more pronounced in the predominantly Ca(2+)(i)-dependent triggered arrhythmias.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Aniline Compounds/pharmacology , Animals , Arrhythmias, Cardiac/metabolism , Benzopyrans/pharmacology , Calcium/metabolism , Cardiotonic Agents/pharmacology , Drug Therapy, Combination/methods , Guanidines/pharmacology , Male , Myocardial Reperfusion/methods , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phenyl Ethers/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Sulfones/pharmacology , Tachycardia, Ventricular/drug therapy , Tachycardia, Ventricular/metabolism , Ventricular Fibrillation/drug therapy , Ventricular Fibrillation/metabolism
2.
Br J Pharmacol ; 170(4): 768-78, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23647096

ABSTRACT

BACKGROUND AND PURPOSE: At present there are no small molecule inhibitors that show strong selectivity for the Na(+) /Ca(2+) exchanger (NCX). Hence, we studied the electrophysiological effects of acute administration of ORM-10103, a new NCX inhibitor, on the NCX and L-type Ca(2+) currents and on the formation of early and delayed afterdepolarizations. EXPERIMENTAL APPROACH: Ion currents were recorded by using a voltage clamp technique in canine single ventricular cells, and action potentials were obtained from canine and guinea pig ventricular preparations with the use of microelectrodes. KEY RESULTS: ORM-10103 significantly reduced both the inward and outward NCX currents. Even at a high concentration (10 µM), ORM-10103 did not significantly change the L-type Ca(2+) current or the maximum rate of depolarization (dV/dtmax ), indicative of the fast inward Na(+) current. At 10 µM ORM-10103 did not affect the amplitude or the dV/dtmax of the slow response action potentials recorded from guinea pig papillary muscles, which suggests it had no effect on the L-type Ca(2+) current. ORM-10103 did not influence the Na(+) /K(+) pump or the main K(+) currents of canine ventricular myocytes, except the rapid delayed rectifier K(+) current, which was slightly diminished by the drug at 3 µM. The amplitudes of pharmacologically- induced early and delayed afterdepolarizations were significantly decreased by ORM-10103 (3 and 10 µM) in a concentration-dependent manner. CONCLUSIONS AND IMPLICATIONS: ORM-10103 is a selective inhibitor of the NCX current and can abolish triggered arrhythmias. Hence, it has the potential to be used to prevent arrhythmogenic events.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Benzopyrans/pharmacology , Heart Ventricles/drug effects , Myocytes, Cardiac/drug effects , Pyridines/pharmacology , Sodium-Calcium Exchanger/antagonists & inhibitors , Action Potentials , Animals , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Dogs , Dose-Response Relationship, Drug , Female , Guinea Pigs , Heart Ventricles/metabolism , Male , Myocytes, Cardiac/metabolism , Papillary Muscles/drug effects , Papillary Muscles/metabolism , Potassium/metabolism , Purkinje Fibers/drug effects , Purkinje Fibers/metabolism , Sodium/metabolism , Sodium-Calcium Exchanger/metabolism , Time Factors
3.
Acta Physiol (Oxf) ; 208(4): 410-21, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23710679

ABSTRACT

AIM: We investigated whether preconditioning with caloric restriction (CR) ameliorates kidney ischaemia/reperfusion (I/R) injury and whether the salutary effects of CR are mediated through enhanced autophagy and/or activation of key metabolic sensors SIRT1, AMP-kinase and PGC-1α. METHODS: Six- to seven-week-old Wistar rats were divided into three groups: (i) sham-operated group; (ii) I/R group (40-min ischaemia followed by 24 h of reperfusion); and (iii) I/R group kept under CR (energy intake 70%) for 2 weeks before surgery. In additional experiments, sirtinol and 3-methyladenine (3-MA) were used as inhibitors of SIRT1 and autophagy respectively. Renal function was measured, and acute tubular damage and nitrotyrosine expression were scored. Kidney adenosine monophosphate-activated kinase (AMPK), SIRT1, eNOS, PGC-1α and LC-3B expressions were measured. RESULTS: Caloric restriction improved renal function, protected against the development of acute tubular necrosis and attenuated I/R-induced nitrosative stress. Kidney I/R injury decreased eNOS and PGC-1α expression, inhibit autophagy and increased SIRT1 and AMPK expressions by 2.6- and fourfold respectively. However, phosphorylation level of AMPK was decreased. As compared with I/R injury group, CR further increased kidney SIRT1 expression by 1.8-fold, promoted autophagy and counteracted I/R-induced decreases in the expression of eNOS and PGC-1α. 3-MA abolished the renoprotective effects of CR, whereas sirtinol did not influence renal function in CR rats with I/R injury. CONCLUSIONS: Caloric restriction ameliorates acute kidney I/R injury through enhanced autophagy and counteraction of I/R-induced decreases in the renal expression of eNOS and PGC-1α.


Subject(s)
Caloric Restriction , Gene Expression Regulation/physiology , Kidney/blood supply , Nitric Oxide Synthase Type III/metabolism , RNA-Binding Proteins/metabolism , Reperfusion Injury/metabolism , Transcription Factors/metabolism , Animals , Autophagy/physiology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Inflammation/metabolism , Kidney/pathology , Kidney/physiology , Male , Nitric Oxide Synthase Type III/genetics , Organ Size , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA-Binding Proteins/genetics , Rats , Rats, Wistar , Stress, Physiological , Transcription Factors/genetics
4.
Br J Pharmacol ; 166(6): 1905-15, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22324445

ABSTRACT

BACKGROUND AND PURPOSE Renal ischaemia/reperfusion (RI/R) injury is a major cause of acute kidney injury (AKI) and an important determinant of long-term kidney dysfunction. AMP-kinase and histone deacetylase sirtuin 1 (SIRT1) regulate cellular metabolism and are activated during hypoxia. We investigated whether AMP-kinase activator AICAR (5-amino-4-imidazolecarboxamide riboside-1-ß-D-ribofuranoside) ameliorates RI/R injury and whether SIRT1 is involved in the pathogenesis. EXPERIMENTAL APPROACH Eight-week-old Sprague Dawley rats were divided into five groups: (i) sham-operated group; (ii) I/R group (40 min bilateral ischaemia followed by 24 h of reperfusion; (iii) I/R group + AICAR 50 mg·kg(-1) i.v. given 60 min before operation; (iv). I/R group + AICAR 160 mg·kg(-1) i.v; (v) I/R group + AICAR 500 mg·kg(-1) i.v. Serum creatinine and urea levels were measured. Acute tubular necrosis (ATN), monocyte/macrophage infiltration and nitrotyrosine expression were scored. Kidney AMP-activated protein kinase (AMPK) and SIRT1 expressions were measured. KEY RESULTS Highest dose of AICAR decreased serum creatinine and urea levels, attenuated I/R injury-induced nitrosative stress and monocyte/macrophage infiltration, and ameliorated the development of ATN. Kidney I/R injury was associated with decreased AMPK phosphorylation and a fivefold increase in kidney SIRT1 expression. AICAR increased pAMPK/AMPK ratio and prevented the I/R-induced increase in renal SIRT1 expression. CONCLUSIONS AND IMPLICATIONS AICAR protects against the development of ATN after kidney I/R injury. Activators of kidney AMP kinase may thus represent a novel therapeutic approach to patients susceptible to AKI and to those undergoing kidney transplantation. The present study also suggests a role for SIRT1 in the pathogenesis of RI/R injury.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Acute Kidney Injury/drug therapy , Aminoimidazole Carboxamide/analogs & derivatives , Protective Agents/therapeutic use , Reperfusion Injury/drug therapy , Ribonucleotides/therapeutic use , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Aminoimidazole Carboxamide/pharmacology , Aminoimidazole Carboxamide/therapeutic use , Animals , Creatinine/blood , Male , Necrosis/drug therapy , Necrosis/metabolism , Necrosis/pathology , Protective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Ribonucleotides/pharmacology , Sirtuin 1/metabolism , Urea/blood
5.
J Physiol Pharmacol ; 62(3): 275-85, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21893687

ABSTRACT

Hypertension is the main risk factor for left ventricular hypertrophy and development of diastolic heart failure. There is no yet treatment, which can effectively reduce mortality in patients suffering from heart failure with preserved systolic function. We tested whether the calcium sensitizer levosimendan and the AT1-receptor antagonist valsartan could protect from salt-induced hypertension, cardiovascular mortality and heart failure in Dahl/Rapp salt-sensitive rats fed for 7 weeks with a high salt diet (8% NaCl). Levosimendan (1 mg/kg/day via drinking water) and valsartan (30 mg/kg in the food) monotherapies and their combination prevented mortality in Dahl/Rapp rats. The drug combination evoked an additive effect on blood pressure, cardiac hypertrophy, cardiomyocyte cross-sectional area, target organ damage and myocardial ANP mRNA expression. There was a close correlation between systolic blood pressure and cardiac hypertrophy, cardiac and renal damage. As compared to Dahl/Rapp controls kept on low-salt diet (NaCl 0.3%). The high salt rats exhibited impaired diastolic relaxation as assessed by isovolumic relaxation time. Levosimendan alone and in combination with valsartan, improved diastolic relaxation without significantly improving systolic function. Our findings are evidence for an additive effect between levosimendan and valsartan on blood pressure and a blood pressure-dependent protection against the development of salt-induced target organ damage. The present study also demonstrates that levosimendan, alone or in combination with valsartan, can correct diastolic dysfunction induced by salt-dependent hypertension.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Cardiovascular Diseases/mortality , Hydrazones/pharmacology , Hypertension/drug therapy , Hypertension/physiopathology , Pyridazines/pharmacology , Tetrazoles/pharmacology , Valine/analogs & derivatives , Vasodilator Agents/pharmacology , Animals , Antihypertensive Agents/pharmacology , Atrial Natriuretic Factor/biosynthesis , Atrial Natriuretic Factor/genetics , Blood Pressure/physiology , Cardiomegaly/complications , Cardiomegaly/diagnosis , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/etiology , Cardiovascular Diseases/pathology , Diet, Sodium-Restricted , Echocardiography , Heart/anatomy & histology , Heart Failure/complications , Heart Failure/physiopathology , Heart Rate , Kidney/anatomy & histology , Kidney/metabolism , Kidney/physiopathology , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Dahl , Simendan , Sodium Chloride, Dietary/administration & dosage , Sodium, Dietary/administration & dosage , Valine/pharmacology , Valsartan , Ventricular Remodeling
6.
J Physiol Pharmacol ; 60(3): 41-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19826180

ABSTRACT

Calcium-sensitizing agents have been shown to improve cardiac function in patients suffering from acute decompensated heart failure, however, their long-term effects on cardiac remodeling and cardiovascular mortality are still largely unknown. In the present study we tested the hypothesis whether OR-1896, an active and long-lasting metabolite of calcium sensitizer levosimendan, prevents cardiovascular mortality and hypertension-induced myocardial remodelling in salt-sensitive Dahl/Rapp rats. OR-1896 was given orally to Dahl/Rapp SS rats on high-salt diet (NaCl 7% w/w) for 7 weeks at two different doses (0.5 and 0.05 mg/kg). OR-1896 prevented salt-induced cardiovascular mortality (survival rate 75 % in OR-1896 treated groups vs 38 % in untreated controls, p<0.01), ameliorated cardiac hypertrophy and improved systolic functions of the heart without major influence on systemic blood pressure. OR-1896 also ameliorated salt-induced increase in cardiac ANP mRNA expression and plasma BNP level. Salt-induced cardiac remodelling was associated with 4-fold increase in cardiac p16(INK4a) mRNA expression, a marker of cellular senescence. OR-1896 dose-dependently ameliorated cardiomyocyte senescence. Our findings suggest a therapeutic role for OR-1896 in the prevention of cardiac remodelling in salt-sensitive forms of hypertension. The present study also underscores the importance of cellular senescence in the pathogenesis of salt-induced hypertensive heart disease.


Subject(s)
Acetamides/therapeutic use , Calcium/physiology , Cardiomegaly/prevention & control , Cardiotonic Agents/therapeutic use , Hypertension/prevention & control , Pyridazines/therapeutic use , Ventricular Remodeling/drug effects , Acetamides/metabolism , Acetamides/pharmacology , Albuminuria/urine , Animals , Blood Pressure/drug effects , Calcium/metabolism , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiotonic Agents/metabolism , Cardiotonic Agents/pharmacology , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Dose-Response Relationship, Drug , Echocardiography , Heart Rate/drug effects , Hypertension/etiology , Hypertension/metabolism , Hypertension/physiopathology , Kidney/pathology , Male , Myocardium/metabolism , Myocardium/pathology , Pyridazines/metabolism , Pyridazines/pharmacology , Rats , Rats, Inbred Dahl , Sodium Chloride, Dietary/adverse effects , Survival Analysis
7.
Eur J Clin Invest ; 39(10): 876-82, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19772522

ABSTRACT

BACKGROUND: Acute heart failure is a potentially fatal manifestation of viral myocarditis. Development of myocardial damage in myocarditis involves cardiomyocyte apoptosis. Levosimendan is a novel calcium sensitizing inotropic agent with anti-apoptotic properties. We studied the feasibility of inotropic treatment with levosimendan and its effects on apoptosis in experimental acute heart failure caused by coxsackievirus myocarditis. MATERIALS AND METHODS: Adolescent BALB/c mice were infected with myocarditic Woodruff variant of coxsackievirus B3 (2 x 10(4) plaque-forming units). Mice were randomized into those receiving levosimendan 0.33 mg kg(-1) (total dose 1 mg kg(-1) day(-1)) (n = 20) or vehicle (n = 19) given orally by gauge three times a day for 7 days after infection. Left ventricular function was evaluated by transthoracic echocardiography and the mice were euthanized on day 7. Histopathology, amount of virus in the heart (virus titration assay) and cardiomyocyte apoptosis (TUNEL assay) were studied. Uninfected untreated control mice were also studied. RESULTS: Infection resulted in histopathologically severe myocarditis and significant impairment of left ventricular function. Levosimendan treatment significantly improved ventricular function (fractional shortening 0.32 +/- 0.04 vs. 0.23 +/- 0.05, P = 0.005; contractility 0.60 +/- 0.12 vs. 0.39 +/- 0.14, P = 0.007 and myocardial performance index 0.36 +/- 0.06 vs. 0.62 +/- 0.15, P < 0.0001) compared with vehicle. Levosimendan also reduced cardiomyocyte apoptosis (0.26 +/- 0.08% vs. 0.44 +/- 0.15% in vehicle, P = 0.008), but did not have an effect on areas of myocardial necrosis or inflammation, or the amount of virus in the heart. Levosimendan treatment did not affect mortality (total mortality 63%). CONCLUSIONS; Levosimendan improves ventricular function and inhibits cardiomyocyte apoptosis; therefore, it is suggested as a potentially feasible therapy in acute heart failure caused by viral myocarditis.


Subject(s)
Cardiotonic Agents/pharmacology , Coxsackievirus Infections/pathology , Heart Failure/pathology , Hydrazones/pharmacology , Myocarditis/pathology , Myocardium/pathology , Pyridazines/pharmacology , Animals , Coxsackievirus Infections/drug therapy , Heart Failure/drug therapy , Male , Mice , Mice, Inbred BALB C , Myocarditis/drug therapy , Myocarditis/virology , Myocytes, Cardiac/pathology , Simendan , Ventricular Function, Left/drug effects
8.
Br J Pharmacol ; 150(7): 851-61, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17325658

ABSTRACT

BACKGROUND AND PURPOSE: Progression of heart failure in hypertensive Dahl rats is associated with cardiac remodeling and increased cardiomyocyte apoptosis. This study was conducted to study whether treatment with a novel inotropic vasodilator compound, levosimendan, could prevent hypertension-induced cardiac remodeling and cardiomyocyte apoptosis. EXPERIMENTAL APPROACH: 6-week-old salt-sensitive Dahl/Rapp rats received levosimendan (0.3 mg kg(-1) and 3 mg kg(-1) via drinking fluid) and high salt diet (NaCl 7%) for 7 weeks, Dahl/Rapp rats on low-salt diet served as controls. Blood pressure, cardiac functions by echocardiography, cardiomyocyte apoptosis by TUNEL technique, tissue morphology, myocardial expression of calcium cycling proteins, and markers of neurohumoral activation were determined. KEY RESULTS: Untreated Dahl/Rapp rats on high salt diet developed severe hypertension, cardiac hypertrophy and moderate systolic dysfunction. 38% of Dahl/Rapp rats (9/24) survived the 7-week-follow-up period. Cardiomyocyte apoptosis was increased by 6-fold during high salt diet. Levosimendan improved survival (survival rates in low- and high-dose levosimendan groups 12/12 and 9/12, p<0.001 and p=0.05, respectively), increased cardiac function, and ameliorated cardiac hypertrophy. Levosimendan dose-dependently prevented cardiomyocyte apoptosis. Levosimendan normalized salt-induced increased expression of natriuretic peptide, and decreased urinary noradrenaline excretion. Levosimendan also corrected salt-induced decreases in myocardial SERCA2a protein expression and myocardial SERCA2a/NCX-ratio. CONCLUSIONS AND IMPLICATIONS: Improved survival by the novel inotropic vasodilator levosimendan in hypertensive Dahl/Rapp rats is mediated, at least in part, by amelioration of hypertension-induced cardiac remodeling and cardiomyocyte apoptosis.


Subject(s)
Hydrazones/pharmacology , Hypertension/drug therapy , Myocytes, Cardiac/drug effects , Pyridazines/pharmacology , Vasodilator Agents/pharmacology , Ventricular Remodeling/drug effects , Animals , Apoptosis/drug effects , Atrial Natriuretic Factor/genetics , Hypertension/metabolism , Male , Myocardium/metabolism , Myocardium/pathology , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/genetics , Norepinephrine/urine , Osteopontin/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Dahl , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Simendan , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism
9.
Eur J Pharmacol ; 419(2-3): 243-8, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11426847

ABSTRACT

This study compared the effects of simendan, a calcium sensitizer, with those of milrinone and enalapril on survival of rats with healed myocardial infarction. Seven days after ligation-induced myocardial infarction, the rats were randomized to control, milrinone, enalapril, or simendan groups. All compounds were administered via the drinking water for 312 days, at which time there was 80% mortality in the control group--the study's primary endpoint. The infarct sizes were similar across all groups. At endpoint, the mortality rates were: 63% (milrinone), 56% (enalapril) and 53% (simendan); the risk reductions were 25% (P = 0.04 vs. control) and 28% (P = 0.02 vs. control) with enalapril and simendan, respectively. Milrinone had no statistically significant effect on the survival rate. These findings suggest that, like enalapril, simendan improved survival in rats with healed myocardial infarction.


Subject(s)
Cardiotonic Agents/therapeutic use , Enalapril/therapeutic use , Hydrazones/therapeutic use , Myocardial Infarction/drug therapy , Pyridazines/therapeutic use , Animals , Antihypertensive Agents/therapeutic use , Male , Milrinone/therapeutic use , Myocardial Infarction/pathology , Rats , Rats, Wistar , Simendan
10.
J Cardiovasc Pharmacol ; 37(4): 367-74, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11300649

ABSTRACT

Levosimendan, a novel calcium sensitizer developed for the treatment of acute heart failure, is an inodilator that increases coronary flow. Because it was recently shown that levosimendan stimulates potassium current through K(ATP) channels in isolated rat arterial cells, our aim was to assess whether the levosimendan-induced increase in coronary flow is due to the opening of the K(ATP) channels in coronary smooth muscle. The effect of levosimendan on the diastolic coronary flow velocity (DCFV) was measured in the Langendorff perfused spontaneously beating guinea-pig heart in the absence and presence of glibenclamide. Pinacidil was used as a reference compound, and the protein kinase C inhibitor bisindolylmaleimide was used to study the dilatory effect of levosimendan when the K(ATP) channels in smooth muscle are not inhibited by PKC-dependent phosphorylation. Levosimendan (0.01-1 microM) increased DCFV concentration-dependently and was noncompetitively antagonized by 0.1 microM glibenclamide, whereas pinacidil was inhibited competitively by glibenclamide. In the presence of glibenclamide the positive inotropic and chronotropic effects of levosimendan were unaltered. The effect of bisindolylmaleimide and levosimendan on DCFV was additive. The results indicate that levosimendan induced coronary vasodilation through the opening of the K(ATP) channels. Levosimendan and pinacidil probably have different binding sites on the K(ATP) channels. The additive effect of bisindolylmaleimide and levosimendan on the increase of DCFV suggests that the latter binds to the unphosphorylated form of the channel.


Subject(s)
Coronary Circulation/drug effects , Heart/drug effects , Hydrazones/pharmacology , Potassium Channels/metabolism , Pyridazines/pharmacology , Vasodilator Agents/pharmacology , Animals , Coronary Circulation/physiology , Female , Glyburide/pharmacology , Guinea Pigs , Heart/physiology , Heart Rate/drug effects , Heart Rate/physiology , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Male , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocardium/metabolism , Pinacidil/pharmacology , Simendan
11.
J Mol Cell Cardiol ; 32(3): 479-91, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10731446

ABSTRACT

Levosimendan, an inodilatory drug discovered using troponin C as a target protein, has a cardiac effect deriving from the calcium sensitization of contractile proteins. The aim of this study was to give further evidence that levosimendan binds to cardiac troponin C and that the binding involves amino acid residues on helixepsilon of the N-terminal domain of this calcium-binding protein. Nine organic molecules, obtained by chemical modification of levosimendan, were tested both for their calcium-dependent binding to troponin C and troponin complex affinity HPLC columns, and for their ability to increase the calcium sensitivity of myofilaments in cardiac skinned fibers. A good correlation between the calcium sensitization and the calcium-dependent binding to troponin complex (r=0.90) and to cardiac troponin C (r=0.91) for the analogs of levosimendan was shown. In addition, the effect of levosimendan on the calcium-induced conformational changes in native and point-mutated cTnC was studied. Cys84-->Ser, Asp87-->Lys and Asp88-->Ala point-mutated cTnC were shown to maintain a high affinity to calcium, but their Ca(2+)titration curves were not influenced by levosimendan as for the native protein. Finally, it was demonstrated that the NMR chemical shifts of the terminal methyl groups of Met47, Met81, and Met85 on calcium-saturated cTnC were changed after addition of levosimendan in water solution at pH 7.4. This effect was not seen when adding an analog of levosimendan, which did not bind to the troponin C affinity HPLC column and did not increase the calcium-induced tension in cardiac skinned fibers.


Subject(s)
Calcium/metabolism , Hydrazones/metabolism , Pyridazines/metabolism , Troponin C/metabolism , Vasodilator Agents/metabolism , Chromatography, Affinity/methods , Chromatography, High Pressure Liquid/methods , Humans , Hydrazones/chemistry , Molecular Structure , Myocardium/metabolism , Nuclear Magnetic Resonance, Biomolecular , Pyridazines/chemistry , Simendan , Structure-Activity Relationship , Troponin C/genetics , Vasodilator Agents/chemistry
12.
Cardiovasc Res ; 34(3): 536-46, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9231037

ABSTRACT

OBJECTIVE: The role of phosphodiesterase III inhibition and calcium sensitization in the cardiac actions of levosimendan, (R)-[[4-(1,4,5, 6-tetrahydro-4-methyl-6-oxo-3-pyridazinyl)phenyl]hydrazono]propane dinitrile, was studied. METHODS: Various heart preparations were used to investigate positive inotropy, chromotropy, coronary flow and calcium sensitivity of contractile proteins. The cAMP- and cGMP-dependent protein kinases (PKA and PKG) were inhibited by KT5720 and KT5823, respectively. Furthermore, the synthesis of cAMP was stimulated by forskolin and increased phosphorylation of troponin I was induced by isoprenaline. RESULTS: In Langendorff guinea-pig heart, levosimendan (0.01-1 microM) and milrinone (0.1-10 microM) increased the left ventricular systolic peak pressure almost to the same extent. In the presence of KT5720 (1 microM) milrinone was devoid of positive inotropic activity. In contrast, KT5720 did not antagonize the inotropic effect of levosimendan at < or = 0.03 microM (-up to the EC50 of levosimendan). The effects of levosimendan and milrinone on heart rate and coronary flow were not affected by KT5720. The PKG inhibitor, KT5823 (1 microM), on the other hand, potentiated the levosimendan-induced increase in coronary flow while it had no effect on the increase induced by milrinone. The mechanical parameters were not affected by KT5823. In the papillary muscle, the positive inotropic effect of milrinone but not that of levosimendan was potentiated by forskolin (0.1 microM). In contrast to milrinone, the positive inotropy by levosimendan was decreased by isoprenaline pretreatment (0.1 microM; 3 min). In line with this, the calcium-sensitizing effect of levosimendan was decreased in skinned fibers prepared from isoprenaline-treated hearts. CONCLUSIONS: Our results indicate that the cardiac effects of levosimendan at its therapeutically relevant concentrations were not mediated through PKA or PKG and its positive inotropy is therefore most probably due to the previously reported troponin-C-mediated calcium sensitization of contractile proteins.


Subject(s)
Calcium/metabolism , Carbazoles , Hydrazones/pharmacology , Myocardial Contraction/drug effects , Myocardium/metabolism , Phosphodiesterase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Pyridazines/pharmacology , Alkaloids/pharmacology , Animals , Cardiotonic Agents/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Guinea Pigs , In Vitro Techniques , Indoles/pharmacology , Milrinone , Papillary Muscles/drug effects , Perfusion , Pyridones/pharmacology , Pyrroles/pharmacology , Simendan , Stimulation, Chemical
13.
J Mol Cell Cardiol ; 27(10): 2155-65, 1995 Oct.
Article in English | MEDLINE | ID: mdl-8576932

ABSTRACT

The effects of various calcium sensitizers on myosin-actin crossbridge kinetics were evaluated in intact, paced guinea-pig papillary muscle by analysing the velocity of the development of isometric tension (dT/dt) in detail. The effect on association (the whole sequence of events from troponin onward) and dissociation rates of crossbridges was estimated from the rising phase and from the early decay phase of the normalized dT/dt curve. Levosimendan, a calcium sensitizer acting through troponin C, accelerated the proportional association rate and decelerated the dissociation rate of crossbridges. The effect of levosimendan on crossbridge kinetics occurred before the peak twitch tension was achieved. Thus, the compound did not change the actual relaxation phase of twitch tension. Since the effect on the association was more pronounced than on the dissociation of crossbridges, levosimendan shifted the entire twitch tension curve to the left. Based on the dissociation rate analysis levosimendan seems to act preferentially as a calcium sensitizer at low concentrations. At high concentrations the phosphodiesterase III (PDE III) inhibitory properties of levosimendan modulated its effect on the early relaxation processes. In contrast, PDE III inhibition is probably the primary mechanism of action for MCI-154. Pimobendan, and EMD 53998 at low concentrations, whereas their direct effects on crossbridge kinetics contributed to the positive inotropic action at high concentrations. The calcium sensitizing mechanisms of these compounds seemed to be based almost exclusively on the decelerating effect on dissociation of crossbridges.


Subject(s)
Calcium/pharmacology , Cardiotonic Agents/pharmacology , Hydrazones/pharmacology , Myocardial Contraction/drug effects , Phosphodiesterase Inhibitors/pharmacology , Pyridazines/pharmacology , Troponin/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Actomyosin/metabolism , Animals , Cyclic Nucleotide Phosphodiesterases, Type 3 , Guinea Pigs , Isometric Contraction/drug effects , Papillary Muscles/chemistry , Papillary Muscles/drug effects , Quinolines/pharmacology , Simendan , Thiadiazines/pharmacology , Troponin C
14.
J Mol Cell Cardiol ; 27(9): 1859-66, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8523447

ABSTRACT

The role of cardiac troponin C (cTnC) as a target protein for the calcium sensitization by levosimendan, pimobendan, MCI-154 and EMD 53998 was evaluated using purified recombinant human cTnC. For determination of calcium- and magnesium-dependent binding of the compounds to cTnC a new type of cTnC-HPLAC column was used. Furthermore, dansylated cTnC was utilized to study the effect of the calcium sensitizing compounds on calcium-induced conformation of cTnC. Only levosimendan showed calcium-dependent and to a lesser extent magnesium-dependent retention in the cTnC column. The findings indicate that levosimendan binds both to the N-terminal and C-terminal domains of cTnC. In agreement with this, only levosimendan shifted the calcium-induced fluorescence curve of dansylated cTnC to the left. In the control experiments Ca50 and KCa2+ were calculated to be 2.73 microM and 4 x 10(5) M-1, respectively. Levosimendan at 3 microM decreased the value of Ca50 to 1.19 microM. In conclusion, it is suggested that the mechanism of calcium sensitizing effect of levosimendan, unlike that of the other calcium sensitizers, is based on calcium-dependent binding to the N-terminal domain of cTnC. This is proposed to amplify the trigger of contraction induced by cTnC in the cardiac muscle.


Subject(s)
Calcium/metabolism , Hydrazones/metabolism , Myocardium/metabolism , Pyridazines/metabolism , Troponin/metabolism , Cardiotonic Agents/pharmacology , Chromatography, Affinity , Humans , Hydrazones/pharmacology , Protein Binding , Protein Conformation/drug effects , Pyridazines/pharmacology , Recombinant Proteins/metabolism , Simendan , Troponin C
15.
J Cardiovasc Pharmacol ; 25(5): 794-801, 1995 May.
Article in English | MEDLINE | ID: mdl-7630157

ABSTRACT

Levosimendan is a novel positive inotropic drug targeted to increase contraction force of the heart through its calcium-dependent binding to troponin C (cTnC). We investigated the calcium-sensitizing effect of levosimendan on contractile proteins as well as its positive inotropic and lusitropic effects in paced guinea pig papillary muscle. We also studied the effect on energy consumption of myosin-actin crossbridges in a myosin ATPase assay. The calcium sensitization induced by levosimendan in fibers skinned with saponin was dependent on the perforation velocity of cell membranes. Levosimendan was almost ineffective in slowly perforated fibers, but was the most potent calcium sensitizer in fibers with rapidly perforated cells. The perforation-dependent calcium sensitization was probably due to changes in phosphorylation state of contractile proteins during the slow dissection of fibers. It is noteworthy that the calcium-sensitizing effect of levosimendan was not affected by acidic pH. Levosimendan at therapeutically relevant (0.3-10 microM) concentrations markedly increased calcium sensitivity both at pH 6.7 and 7.0, being more potent than EMD 53998, pimobendan, and MCI-154. The lack of effect of levosimendan on maximum tension supports the hypothesis that levosimendan increases calcium sensitivity through its action on cTnC. Unlike EMD 53998, levosimendan did not increase myosin ATPase activity, indicating that it did not increase the cycling rate of myosinactin crossbridges. In paced papillary muscles, levosimendan induced positive inotropic effect without changing relaxation time. Thus, levosimendan was devoid of the main negative factors described for calcium sensitizers.


Subject(s)
Calcium/metabolism , Cardiotonic Agents/pharmacology , Hydrazones/pharmacology , Myocardial Contraction/drug effects , Pyridazines/pharmacology , Troponin/pharmacology , Adenosine Triphosphatases/metabolism , Animals , Cardiac Pacing, Artificial , Cell Membrane/drug effects , Female , Guinea Pigs , Hydrogen-Ion Concentration , Male , Myofibrils/enzymology , Papillary Muscles/cytology , Papillary Muscles/drug effects , Papillary Muscles/metabolism , Quinolines/pharmacology , Simendan , Thiadiazines/pharmacology , Troponin C , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL