Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Naunyn Schmiedebergs Arch Pharmacol ; 394(2): 291-298, 2021 02.
Article in English | MEDLINE | ID: mdl-32949251

ABSTRACT

Atrial fibrillation (AF)-associated remodeling includes contractile dysfunction whose reasons are only partially resolved. Serotonin (5-HT) increases contractile force and causes arrhythmias in atrial trabeculae from patients in sinus rhythm (SR). In persistent atrial fibrillation (peAF), the force responses to 5-HT are blunted and arrhythmic effects are abolished. Since force but not arrhythmic responses to 5-HT in peAF could be restored by PDE3 + PDE4 inhibition, we sought to perform real-time measurements of cAMP to understand whether peAF alters PDE3 + PDE4-mediated compartmentation of 5-HT4 receptor-cAMP responses. Isolated human atrial myocytes from patients in SR, with paroxysmal AF (paAF) or peAF, were adenovirally transduced to express the FRET-based cAMP sensor Epac1-camps. Forty-eight hours later, cAMP responses to 5-HT (100 µM) were measured in the absence or concomitant presence of the PDE3 inhibitor cilostamide (0.3 µM) and the PDE4 inhibitor rolipram (1 µM). We successfully established real-time cAMP imaging in AF myocytes. 5-HT increased cAMP in SR, paAF, and peAF, but in line with previous findings on contractility, this increase was considerably smaller in peAF than in SR or paAF. The maximal cAMP response to forskolin (10 µM) was preserved in all groups. The diminished cAMP response to 5-HT in peAF was recovered by preincubation with cilostamide + rolipram. We uncovered a significantly diminished cAMP response to 5-HT4 receptor stimulation which may explain the blunted 5-HT inotropic responses observed in peAF. Since both cAMP and force responses but not arrhythmic responses were recovered after concomitant inhibition of PDE3 + PDE4, they might be regulated in different subcellular microdomains.


Subject(s)
Atrial Fibrillation/metabolism , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Myocytes, Cardiac/metabolism , Receptors, Serotonin, 5-HT4/metabolism , Aged , Female , Humans , Male , Middle Aged
2.
Naunyn Schmiedebergs Arch Pharmacol ; 391(6): 571-585, 2018 06.
Article in English | MEDLINE | ID: mdl-29556684

ABSTRACT

Myocardial PDE2 activity increases in terminal human heart failure and after isoprenaline infusion in rat heart. PDE2 inhibitors do not potentiate the murine sinoatrial tachycardia produced by noradrenaline. We investigated whether isoprenaline infusion induces PDE2 to decrease the chronotropic and inotropic effects of catecholamines in rat heart. Sprague-Dawley rats were infused with isoprenaline (2.4 mg kg-1 day-1) for 3 days. We used spontaneously beating right atria, paced right ventricular strips and left ventricular papillary muscles. The effects of the PDE2 inhibitors EHNA (10 µM) and Bay 60-7550 (0.1-1 µM) were investigated on the cardiostimulation produced by noradrenaline (ICI118551 50 nM present to block ß2-adrenoceptors) and adrenaline (CGP20712A 300 nM present to block ß1-adrenoceptors). Hydrolysis of cAMP by PDE2 was measured by radioenzyme assay. Bay 60-7550 but not EHNA increased sinoatrial beating. A stable tachycardia elicited by noradrenaline (10 nM) or adrenaline (1 µM) was not increased by the PDE2 inhibitors. Isoprenaline infusion increased the hydrolytic PDE2 activity threefold in left ventricle, reduced the chronotropic and inotropic effects and potency of noradrenaline and abolished the effects of adrenaline. The potency of the catecholamines was not increased by the PDE2 inhibitors. Neither EHNA nor Bay 60-7550 potentiated the effects of the catecholamines. Rat PDE2 decreased basal sinoatrial beating but did not reduce the sinoatrial tachycardia or increases of ventricular force mediated through ß1- and ß2-adrenoceptors. The ß-adrenoceptor desensitization induced by the isoprenaline infusion was not reversed by the PDE2 inhibitors despite the increased hydrolysis of cAMP by PDE2.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 2/physiology , Heart/drug effects , Isoproterenol/pharmacology , Receptors, Adrenergic, beta-1/physiology , Receptors, Adrenergic, beta-2/physiology , Animals , Cardiotonic Agents/pharmacology , Epinephrine/pharmacology , Heart/physiology , In Vitro Techniques , Male , Norepinephrine/pharmacology , Rats, Sprague-Dawley , Tachycardia/chemically induced
3.
Br J Pharmacol ; 174(16): 2706-2715, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28574581

ABSTRACT

BACKGROUND AND PURPOSE: Mirabegron has been classified as a ß3 -adrenoceptor agonist approved for overactive bladder syndrome. We investigated possible cardiac effects of mirabegron in the absence or presence of ß-adrenoceptor subtype antagonists. In view of its phenylethanolamine structure, we investigated whether mirabegron has indirect sympathomimetic activity by using neuronal uptake blockers. EXPERIMENTAL APPROACH: Right atrial trabeculae, from non-failing hearts, were paced and contractile force measured at 37°C. Single concentrations of mirabegron were added in the absence or presence of the phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine (IBMX), ß3 (L-748,337), ß1 (CGP 20712A), ß2 (ICI 118,551) -adrenoceptor antagonists, neuronal uptake inhibitors desipramine or phenoxybenzamine. KEY RESULTS: Mirabegron significantly increased contractile force in human right atrium (1 µM, 7.6 ± 2.6%, n = 7; 10 µM, 10.2 ± 1.5%, n = 22 compared with (-)-isoprenaline P < 0.05). In the presence of IBMX, mirabegron (10 µM) caused a greater contraction. L-748,337 (100 nM) had no effect on the increase in contractile force caused by mirabegron (10 µM). In contrast, mirabegron (10 µM) reduced contractile force in the presence of CGP 20712A, which was not affected by L-748,337 (100 nM) or ICI 118,551 (50 nM). Mirabegron (10 µM) also reduced contractile force in the presence of desipramine or phenoxybenzamine. CONCLUSIONS AND IMPLICATIONS: Mirabegron increases human atrial force through ß1 - but not ß3 -adrenoceptors. Desipramine and phenoxybenzamine block neuronal uptake and conceivably prevent mirabegron from releasing noradrenaline. A non-specific cardiodepressant effect is not mediated through ß3 (or ß2 )-adrenoceptors, consistent with lack of ß3 -adrenoceptor function on human atrial contractility.


Subject(s)
Acetanilides/pharmacology , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Atrial Function/drug effects , Heart Atria/drug effects , Thiazoles/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , Aged , Female , Humans , Imidazoles/pharmacology , In Vitro Techniques , Male , Middle Aged , Myocardial Contraction/drug effects , Propanolamines/pharmacology , Receptors, Adrenergic, beta/physiology
4.
Br J Pharmacol ; 173(16): 2478-89, 2016 08.
Article in English | MEDLINE | ID: mdl-27238373

ABSTRACT

BACKGROUND AND PURPOSE: 5-HT increases force and L-type Ca(2) (+) current (ICa,L ) and causes arrhythmias through 5-HT4 receptors in human atrium. In permanent atrial fibrillation (peAF), atrial force responses to 5-HT are blunted, arrhythmias abolished but ICa,L responses only moderately attenuated. We investigated whether, in peAF, this could be due to an increased function of PDE3 and/or PDE4, using the inhibitors cilostamide (300 nM) and rolipram (1 µM) respectively. EXPERIMENTAL APPROACH: Contractile force, arrhythmic contractions and ICa,L were assessed in right atrial trabeculae and myocytes, obtained from patients with sinus rhythm (SR), paroxysmal atrial fibrillation (pAF) and peAF. KEY RESULTS: Maximum force responses to 5-HT were reduced to 15% in peAF, but not in pAF. Cilostamide, but not rolipram, increased both the blunted force responses to 5-HT in peAF and the inotropic potency of 5-HT fourfold to sevenfold in trabeculae of patients with SR, pAF and peAF. Lusitropic responses to 5-HT were not decreased in peAF. Responses of ICa,L to 5-HT did not differ and were unaffected by cilostamide or rolipram in myocytes from patients with SR or peAF. Concurrent cilostamide and rolipram increased 5-HT's propensity to elicit arrhythmias in trabeculae from patients with SR, but not with peAF. CONCLUSIONS AND IMPLICATIONS: PDE3, but not PDE4, reduced inotropic responses to 5-HT in peAF, independently of lusitropy and ICa,L , but PDE3 activity was the same as that in patients with SR and pAF. Atrial remodelling in peAF abolished the facilitation of 5-HT to induce arrhythmias by inhibition of PDE3 plus PDE4.


Subject(s)
Arrhythmias, Cardiac/metabolism , Atrial Fibrillation/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Serotonin/metabolism , Aged , Female , Humans , Male , Phosphodiesterase Inhibitors/pharmacology
5.
Naunyn Schmiedebergs Arch Pharmacol ; 389(2): 177-86, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26531832

ABSTRACT

Phosphodiesterases PDE2, PDE3, and PDE4 are expressed in murine sinoatrial cells. PDE3 and/or PDE4 reduce heart rate but apparently do not influence the tachycardia mediated through sinoatrial ß1- and ß2-adrenoceptors despite the high content of sinoatrial cAMP. The function of PDE2 is, however, uncertain. Prostaglandin PGE1 elicits sinoatrial tachycardia through EP receptors, but the control by phosphodiesterases is unknown. We investigated on spontaneously beating right atria of mice the effects of the PDE2 inhibitors Bay 60-7550 and EHNA on basal beating and the tachycardia produced by noradrenaline (3 nM) and PGE1 (1 µM). Bay 60-7550 (1 µM), but not EHNA (10 µM), increased basal sinoatrial beating. EHNA also failed to produce tachycardia in the presence of the adenosine deaminase inhibitor 2'-deoxycoformycin (10 µM), remaining inconclusive whether PDE2 reduces basal sinoatrial beating. Rolipram (10 µM) and cilostamide (300 nM) caused moderate tachycardia. The tachycardia evoked by Bay 60-7550 was similar in the absence and presence of rolipram. Noradrenaline elicited stable tachycardia that was not increased by Bay 60-7550. A stable tachycardia caused by PGE1 was not increased by the inhibitors of PDE2, PDE3, and PDE4. Unlike PDE3 and PDE4 which reduce murine basal sinoatrial beating, a possible effect of PDE2 needs further research. The stable tachycardia produced by noradrenaline and PGE1, together with the lack potentiation by the inhibitors of PDE2, PDE3, and PDE4, suggests that cAMP generated at the receptor compartments is hardly hydrolyzed by these phophodiesterases. Evidence from human volunteers is consistent with this proposal.


Subject(s)
Alprostadil , Arrhythmia, Sinus/chemically induced , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Heart Rate/drug effects , Norepinephrine , Phosphodiesterase 3 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Tachycardia, Supraventricular/chemically induced , Animals , Arrhythmia, Sinus/enzymology , Arrhythmia, Sinus/physiopathology , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Disease Models, Animal , Hydrolysis , Isolated Heart Preparation , Male , Mice , Phosphodiesterase 3 Inhibitors/toxicity , Phosphodiesterase 4 Inhibitors/toxicity , Receptors, Adrenergic, beta-1/metabolism , Receptors, Prostaglandin E/metabolism , Second Messenger Systems/drug effects , Tachycardia, Supraventricular/enzymology , Tachycardia, Supraventricular/physiopathology , Time Factors
6.
Neurourol Urodyn ; 34(6): 592-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-24820256

ABSTRACT

AIMS: To investigate the role of the mucosa in (-)-isoprenaline-induced relaxation of mouse detrusor muscle and to characterize the ß-adrenoceptor subtypes involved. METHODS: Isolated intact and mucosa-denuded muscle strips from the urinary bladder of male C57BL6 mice were pre-contracted with KCl (40 mM) and were relaxed with increasing concentrations of the ß-adrenoceptor (ß-AR) agonist (-)-isoprenaline and forskolin in the presence and absence of the subtype-selective ß-AR blockers CGP20712A (ß1 -ARs), ICI118,551 (ß2 -ARs), and L748,337 (ß3 -ARs). RESULTS: Force development in response to KCl was larger in mucosa-denuded than in intact preparations and was almost completely relaxed with increasing concentrations of (-)-isoprenaline. Mucosa-denuded muscles were about 10-fold more sensitive to (-)-isoprenaline than intact muscles. CGP20712A did not affect the concentration-response curves (CRCs) to (-)-isoprenaline, ICI118,551 shifted the CRC further to the right in denuded than in intact strips so that the difference between them was abolished. Combined exposure to ß1 -AR and ß2 -AR blocker yielded the same result. L748,337 did not significantly affect the CRC to (-)-isoprenaline but caused additional blockade to ICI118,551 in the presence of intact mucosa. CONCLUSIONS: The mucosa of mouse detrusor strips impairs KCl-induced force development and reduces the sensitivity to ß-AR-induced relaxation. The relaxing response to (-)-isoprenaline as well as the mucosa effect thereupon are mainly mediated by ß2 -ARs. A minor involvement of ß3 -ARs becomes apparent particularly at high (-)-isoprenaline concentrations.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Isoproterenol/pharmacology , Muscle Relaxation/drug effects , Urinary Bladder/drug effects , Adrenergic beta-1 Receptor Antagonists/pharmacology , Aminophenols , Animals , Colforsin/pharmacology , Dose-Response Relationship, Drug , Imidazoles/pharmacology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mucous Membrane/drug effects , Muscle, Smooth/drug effects , Potassium Chloride/pharmacology , Sulfonamides , Vasodilator Agents/pharmacology
7.
Naunyn Schmiedebergs Arch Pharmacol ; 387(7): 629-40, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24668024

ABSTRACT

The ß-blockers carvedilol and metoprolol provide important therapeutic strategies for heart failure treatment. Therapy with metoprolol facilitates the control by phosphodiesterase PDE3, but not PDE4, of inotropic effects of catecholamines in human failing ventricle. However, it is not known whether carvedilol has the same effect. We investigated whether the PDE3-selective inhibitor cilostamide (0.3 µM) or PDE4-selective inhibitor rolipram (1 µM) modified the positive inotropic and lusitropic effects of catecholamines in ventricular myocardium of heart failure patients treated with carvedilol. Right ventricular trabeculae from explanted hearts of nine carvedilol-treated patients with terminal heart failure were paced to contract at 1 Hz. The effects of (-)-noradrenaline, mediated through ß1-adrenoceptors (ß2-adrenoceptors blocked with ICI118551), and (-)-adrenaline, mediated through ß2-adrenoceptors (ß1-adrenoceptors blocked with CGP20712A), were assessed in the absence and presence of the PDE inhibitors. The inotropic potency, estimated from -logEC50s, was unchanged for (-)-noradrenaline but decreased 16-fold for (-)-adrenaline in carvedilol-treated compared to non-ß-blocker-treated patients, consistent with the previously reported ß2-adrenoceptor-selectivity of carvedilol. Cilostamide caused 2- to 3-fold and 10- to 35-fold potentiations of the inotropic and lusitropic effects of (-)-noradrenaline and (-)-adrenaline, respectively, in trabeculae from carvedilol-treated patients. Rolipram did not affect the inotropic and lusitropic potencies of (-)-noradrenaline or (-)-adrenaline. Treatment of heart failure patients with carvedilol induces PDE3 to selectively control the positive inotropic and lusitropic effects mediated through ventricular ß2-adrenoceptors compared to ß1-adrenoceptors. The ß2-adrenoceptor-selectivity of carvedilol may provide protection against ß2-adrenoceptor-mediated ventricular overstimulation in PDE3 inhibitor-treated patients. PDE4 does not control ß1- and ß2-adrenoceptor-mediated inotropic and lusitropic effects in carvedilol-treated patients.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Carbazoles/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 3/physiology , Heart Failure/physiopathology , Propanolamines/pharmacology , Receptors, Adrenergic, beta-1/physiology , Receptors, Adrenergic, beta-2/physiology , Adult , Carvedilol , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Epinephrine/pharmacology , Female , Heart Transplantation , Humans , Male , Middle Aged , Norepinephrine/pharmacology , Phosphodiesterase 3 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Quinolones/pharmacology , Rolipram/pharmacology
8.
BJU Int ; 112(8): 1215-22, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23937341

ABSTRACT

OBJECTIVES: To elucidate the impact of the mucosa on detrusor muscle function by investigating force of contraction under various stimulatory conditions and during subsequent relaxation using catecholamines. PATIENTS AND METHODS: Detrusor tissue was obtained from patients who had undergone cystectomy for bladder cancer and strips of intact or mucosa-denuded muscle were set up for force measurement. Preparations were precontracted with KCl, carbachol or electric-field stimulation (EFS). Precontracted strips were relaxed using increasing concentrations of catecholamines in the absence and presence of the subtype-selective ß-adrenoceptor (AR) blockers CGP 20712A (ß1-ARs), ICI 118,551 (ß2-ARs), and L-748,337 (ß3-ARs). RESULTS: Force development in response to KCl (40 mM), carbachol (1 µM) or EFS was larger in the absence of mucosa than in intact muscle strips. The force of contraction of mucosa-denuded strips with detached urothelium incubated in the same chamber was as low as in intact strips. Noradrenaline relaxed precontracted detrusor strips to a significantly larger extent and at lower concentrations in denuded than in intact strips. CGP 20712A did not affect noradrenaline-induced relaxation of denuded and intact strips, and ICI 118,551 did not influence noradrenaline-induced relaxation in denuded strips, but abolished the difference between denuded and intact strips. The antagonism of the relaxant effects of noradrenaline by L-748,337 was slightly smaller in intact than denuded strips. CONCLUSIONS: The mucosa of human detrusor strips impairs force development when stimulated with KCl, carbachol or EFS. The mucosa also blunts the relaxing effects of catecholamines. The latter effect does not involve the activation of ß1-ARs but only of ß2-ARs, whereas ß3-ARs mediate the relaxation of human detrusor.


Subject(s)
Adrenergic beta-3 Receptor Antagonists/pharmacology , Catecholamines/pharmacology , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Norepinephrine/pharmacology , Receptors, Adrenergic, beta/metabolism , Urinary Bladder/physiopathology , Aged , Dose-Response Relationship, Drug , Female , Humans , Male , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Receptors, Adrenergic, beta-3/metabolism , Urinary Bladder/drug effects , Urothelium/drug effects , Urothelium/physiopathology
9.
Br J Pharmacol ; 169(3): 528-38, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23489141

ABSTRACT

BACKGROUND AND PURPOSE: PDE3 and/or PDE4 control ventricular effects of catecholamines in several species but their relative effects in failing human ventricle are unknown. We investigated whether the PDE3-selective inhibitor cilostamide (0.3-1 µM) or PDE4 inhibitor rolipram (1-10 µM) modified the positive inotropic and lusitropic effects of catecholamines in human failing myocardium. EXPERIMENTAL APPROACH: Right and left ventricular trabeculae from freshly explanted hearts of 5 non-ß-blocker-treated and 15 metoprolol-treated patients with terminal heart failure were paced to contract at 1 Hz. The effects of (-)-noradrenaline, mediated through ß1 adrenoceptors (ß2 adrenoceptors blocked with ICI118551), and (-)-adrenaline, mediated through ß2 adrenoceptors (ß1 adrenoceptors blocked with CGP20712A), were assessed in the absence and presence of PDE inhibitors. Catecholamine potencies were estimated from -logEC50s. KEY RESULTS: Cilostamide did not significantly potentiate the inotropic effects of the catecholamines in non-ß-blocker-treated patients. Cilostamide caused greater potentiation (P = 0.037) of the positive inotropic effects of (-)-adrenaline (0.78 ± 0.12 log units) than (-)-noradrenaline (0.47 ± 0.12 log units) in metoprolol-treated patients. Lusitropic effects of the catecholamines were also potentiated by cilostamide. Rolipram did not affect the inotropic and lusitropic potencies of (-)-noradrenaline or (-)-adrenaline on right and left ventricular trabeculae from metoprolol-treated patients. CONCLUSIONS AND IMPLICATIONS: Metoprolol induces a control by PDE3 of ventricular effects mediated through both ß1 and ß2 adrenoceptors, thereby further reducing sympathetic cardiostimulation in patients with terminal heart failure. Concurrent therapy with a PDE3 blocker and metoprolol could conceivably facilitate cardiostimulation evoked by adrenaline through ß2 adrenoceptors. PDE4 does not appear to reduce inotropic and lusitropic effects of catecholamines in failing human ventricle.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/adverse effects , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Heart Failure/drug therapy , Heart Ventricles/drug effects , Metoprolol/adverse effects , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Adrenergic alpha-Agonists/chemistry , Adrenergic alpha-Agonists/pharmacology , Adrenergic beta-1 Receptor Antagonists/pharmacology , Adrenergic beta-1 Receptor Antagonists/therapeutic use , Adrenergic beta-2 Receptor Antagonists/pharmacology , Adrenergic beta-Agonists/chemistry , Adrenergic beta-Agonists/pharmacology , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/therapeutic use , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Cyclic Nucleotide Phosphodiesterases, Type 3/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Drug Resistance/drug effects , Epinephrine/agonists , Epinephrine/pharmacology , Heart Failure/metabolism , Heart Failure/physiopathology , Heart Failure/surgery , Heart Transplantation , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Humans , In Vitro Techniques , Metoprolol/therapeutic use , Middle Aged , Myocardial Contraction/drug effects , Norepinephrine/agonists , Norepinephrine/pharmacology , Phosphodiesterase 3 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-2/chemistry
11.
Trends Pharmacol Sci ; 32(7): 377-83, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21481950

ABSTRACT

Cyclic AMP (cAMP) steers the generation of basal heart beat in the sinoatrial node. It also induces sinoatrial tachycardia and increased cardiac force, elicited through activation of Gs-protein-coupled receptors (GsPCRs). Phosphodiesterases (PDEs) hydrolyse cAMP. In the heart mainly PDE3 and PDE4 would be expected to limit those functions, and the PDE isoenzymes do indeed reduce basal sinoatrial beating rate and blunt the positive inotropic effects of agonists, mediated by GsPCRs. By contrast, recent evidence shows that GsPCR-mediated sinoatrial tachycardia is not controlled by PDE1-5. A PDE-resistant cAMP pool in sinoatrial cells, generated through activation of GsPCRs, including ß(1)- and ß(2)-adrenoceptors, appears to guarantee unrestrained tachycardia during fight or flight stress.


Subject(s)
Phosphoric Diester Hydrolases/metabolism , Receptors, G-Protein-Coupled/metabolism , Sinoatrial Node/metabolism , Sinoatrial Node/physiopathology , Tachycardia/metabolism , Tachycardia/physiopathology , Animals , Heart Rate/physiology , Humans , Receptors, G-Protein-Coupled/agonists , Tachycardia/enzymology
12.
J Mol Model ; 17(10): 2525-38, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21203789

ABSTRACT

The affinity of the classical ß(2) adrenoceptor-selective inverse agonist ICI118,551 is notoriously lower for porcine ß(2) adrenoceptors (p(2)ßAR) than for human ß(2) adrenoceptors (hß(2)AR) but molecular mechanisms for this difference are still unclear. Homology 3-D models of pß(2)AR can be useful in predicting similarities and differences, which might in turn increase the comparative understanding of ligand interactions with the hß(2)AR. In this work, the pß(2)AR amino acid sequence was used to carry out homology modeling. The selected pß(2)AR 3-D structure was structurally and energetically optimized and used as a model for further theoretical study. The homology model of pß(2)AR has a 3-D structure very similar to the crystal structures of recently studied hß(2)AR. This was also corroborated by sequence identity, RMSD, Ramachandran map, TM-score and docking results. Upon performing molecular docking simulations with the AutoDock4.0.1 program on pß(2)AR, it was found that a set of well-known ß(2)AR ligands reach two distinct binding sites on pß(2)AR. Whereas one of these sites is similar to that reported on the hß(2)AR crystal structure, the other can explain some important experimental observations. Additionally, the theoretical affinity estimated for ICI118,551 closely agrees with affinities estimated from experimental in vitro data. The experimental differences between the human/porcine ß(2)ARs in relation to ligand affinity can in part be elucidated by observations in this molecular modeling study.


Subject(s)
Molecular Dynamics Simulation , Receptors, Adrenergic, beta-2/chemistry , Animals , Binding Sites , Humans , Ligands , Protein Binding , Protein Conformation , Receptors, Adrenergic, beta-2/metabolism , Structural Homology, Protein , Structure-Activity Relationship , Swine/metabolism
13.
Br J Pharmacol ; 162(4): 823-39, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20726983

ABSTRACT

BACKGROUND AND PURPOSE: It has been proposed that BRL37344, SR58611 and CGP12177 activate ß3-adrenoceptors in human atrium to increase contractility and L-type Ca(2+) current (I(Ca-L)). ß3-adrenoceptor agonists are potentially beneficial for the treatment of a variety of diseases but concomitant cardiostimulation would be potentially harmful. It has also been proposed that (-)-CGP12177 activates the low affinity binding site of the ß1-adrenoceptor in human atrium. We therefore used BRL37344, SR58611 and (-)-CGP12177 with selective ß-adrenoceptor subtype antagonists to clarify cardiostimulant ß-adrenoceptor subtypes in human atrium. EXPERIMENTAL APPROACH: Human right atrium was obtained from patients without heart failure undergoing coronary artery bypass or valve surgery. Cardiomyocytes were prepared to test BRL37344, SR58611 and CGP12177 effects on I(Ca-L). Contractile effects were determined on right atrial trabeculae. KEY RESULTS: BRL37344 increased force which was antagonized by blockade of ß1- and ß2-adrenoceptors but not by blockade of ß3-adrenoceptors with ß3-adrenoceptor-selective L-748,337 (1 µM). The ß3-adrenoceptor agonist SR58611 (1 nM-10 µM) did not affect atrial force. BRL37344 and SR58611 did not increase I(Ca-L) at 37°C, but did at 24°C which was prevented by L-748,337. (-)-CGP12177 increased force and I(Ca-L) at both 24°C and 37°C which was prevented by (-)-bupranolol (1-10 µM), but not L-748,337. CONCLUSIONS AND IMPLICATIONS: We conclude that the inotropic responses to BRL37344 are mediated through ß1- and ß2-adrenoceptors. The inotropic and I(Ca-L) responses to (-)-CGP12177 are mediated through the low affinity site ß(1L)-adrenoceptor of the ß1-adrenoceptor. ß3-adrenoceptor-mediated increases in I(Ca-L) are restricted to low temperatures. Human atrial ß3-adrenoceptors do not change contractility and I(Ca-L) at physiological temperature.


Subject(s)
Calcium Channels, L-Type/metabolism , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Aged , Atrial Appendage/cytology , Calcium Signaling/drug effects , Ethanolamines/antagonists & inhibitors , Ethanolamines/pharmacology , Female , Humans , In Vitro Techniques , Kinetics , Male , Middle Aged , Myocytes, Cardiac/metabolism , Propanolamines/antagonists & inhibitors , Propanolamines/pharmacology , Receptors, Adrenergic, beta-3/metabolism , Temperature , Tetrahydronaphthalenes/antagonists & inhibitors , Tetrahydronaphthalenes/pharmacology
14.
Eur J Pharmacol ; 638(1-3): 99-107, 2010 Jul 25.
Article in English | MEDLINE | ID: mdl-20406625

ABSTRACT

The structures of porcine and human beta(2)-adrenoceptors differ but the repercussions for porcine cardiac function are unknown. We investigated the function of porcine beta(2)-adrenoceptors in 3 cardiac regions, sinoatrial node, left atrium and right ventricle of newborn piglets. Both (-)-noradrenaline and (-)-adrenaline caused sinoatrial tachycardia: 60+/-10% and 62+/-7% of the maximum response (E(max)) to (-)-noradrenaline (-logEC(50)=9.0) and (-)-adrenaline (-logEC(50)=7.5) respectively, were resistant to antagonism by the beta(1)-selective CGP20712A (2-hydroxy-5-[2-[[2-hydroxy-3-[4-[1-methyl-4-(trifluoromethyl)-1H-imidazol-2-yl]phenoxy]propyl]amino]ethoxy]-benzamide) (300 nM) but antagonized by beta(2)-selective ICI118551 (erythro(+/-)-[1-(2,3-dihydro-7-methyl-1H-inden-4-yl)oxy]-3-[(1-methylethyl)amino]-2-butanol) (50 nM), consistent with mediation through beta(2)-adrenoceptors. The phosphodiesterase3-selective inhibitor cilostamide and phosphodiesterase4-selective inhibitor rolipram did not affect catecholamine chronotropic potencies. Only small CGP20712A-resistant positive inotropic effects of (-)-adrenaline were detected in the left atria (13+/-2% of E(max)) and ventricular trabeculae (14+/-5% of E(max)). The atrial inotropic responses to (-)-noradrenaline and (-)-adrenaline faded; fades were prevented by rolipram but not cilostamide or concurrent cilostamide+rolipram respectively. (-)-Noradrenaline (ICI118551 present) increased left atrial cAMP levels through beta(1)-adrenoceptors that were markedly enhanced by rolipram but unaffected by cilostamide. Concurrent cilostamide+rolipram uncovered inotropic and cAMP responses to (-)-adrenaline (CGP20712A present). We conclude that sinoatrial beta(2)-adrenoceptors are more important than beta(1)-adrenoceptors in the mediation of tachycardia caused by both (-)-noradrenaline and (-)-adrenaline in the newborn piglet. beta(2)-adrenoceptors have only a minor role in the mediation of left atrial and ventricular inotropic effects of (-)-adrenaline. Catecholamine-evoked tachycardia is not controlled by PDE3 or PDE4. PDE4, but not PDE3, controls the atrial inotropic and cAMP beta(1)-adrenoceptor-mediated responses to (-)-noradrenaline. Both PDE3 and PDE4 blunt left atrial inotropic and cAMP responses to (-)-adrenaline through beta(2)-adrenoceptors.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Receptors, Adrenergic, beta-1/physiology , Receptors, Adrenergic, beta-2/physiology , Tachycardia/physiopathology , Adrenergic Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Animals, Newborn , Cyclic AMP/metabolism , Epinephrine/pharmacology , Heart Atria/drug effects , Heart Atria/metabolism , Heart Ventricles/drug effects , In Vitro Techniques , Norepinephrine/pharmacology , Phosphodiesterase 3 Inhibitors , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Quinolones/pharmacology , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-2/drug effects , Rolipram/pharmacology , Sinoatrial Node/drug effects , Swine , Tachycardia/chemically induced
15.
Naunyn Schmiedebergs Arch Pharmacol ; 380(5): 421-30, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19693491

ABSTRACT

The mammalian heart expresses at least five phosphodiesterases (PDE1-5). Catecholamines produce surges of inotropically relevant cAMP through beta(1)-adrenoceptor stimulation. cAMP is mainly hydrolysed by PDE3 and/or PDE4 thereby blunting contractility. Basal sinoatrial beating rate in mouse, rat, piglet and rabbit sinoatrial cells is reduced by PDE3 and/or PDE4 through hydrolysis of cAMP. However, in rodents, the tachycardia elicited by catecholamines through production of cAMP by beta-adrenoceptor activation is not controlled by PDE3 and PDE4, despite a blunting effect of PDE3 or/and PDE4 on basal sinoatrial beating, but it is unknown whether PDE3 limits catecholamine-evoked tachycardia in the rabbit. Since rabbit sinoatrial cells are an important model for pacemaker research, we investigated whether the positive chronotropic effects of (-)-noradrenaline on spontaneously beating right atria of the rabbit are potentiated by inhibition of PDE3 with cilostamide (300 nM). We also studied the sinoatrial effects of the PDE4 inhibitor rolipram (10 microM) and its influence on the responses to (-)-noradrenaline. For comparison, we investigated the influence of cilostamide and rolipram on the positive inotropic responses to (-)-noradrenaline on rabbit left atria and right ventricular papillary muscles. Cilostamide and concurrent cilostamide + rolipram, but not rolipram alone, increased sinoatrial rate by 15% and 31% of the effect of (-)-isoprenaline (200 microM) but the PDE inhibitors did not significantly change the chronotropic potency of (-)-noradrenaline. In contrast in papillary muscle, the positive inotropic effects of (-)-noradrenaline were potentiated 2.4-, 2.6- and 44-fold by cilostamide, rolipram and concurrent cilostamide + rolipram, respectively. In left atrium, the positive inotropic effects of (-)-noradrenaline were marginally potentiated by cilostamide, as well as potentiated 2.7- and 32-fold by rolipram and by concurrent cilostamide and rolipram respectively. To compare the influence of PDE1-5 on basal sinoatrial rate and (-)-noradrenaline-evoked tachycardia, we investigated on rat right atria the effects of selective inhibitors. The PDE4 inhibitor rolipram and non-selective inhibitor isobutyl-methylxanthine caused tachycardia with -logEC(50)s of 7.2 and 5.0 and E(max) of 18% and 102% of (-)-isoprenaline, respectively. Rolipram did not change the chronotropic potency of (-)-noradrenaline. At high concentrations (10-30 microM), the PDE1, PDE3 and PDE5 inhibitors 8-methoxymethyl-3-isobutyl-1-methylxanthine, cilostamide and sildenafil, respectively, caused marginal tachycardia but did not significantly change the chronotropic potency of (-)-noradrenaline. The PDE2-selective inhibitor erythro-9-[2-hydroxy-3-nonyl]adenine caused marginal bradycardia at 30 microM and tended to reduce the chronotropic potency of (-)-noradrenaline. Rabbit PDE3 reduces basal sinoatrial rate. Although PDE4 only marginally reduces rate, under conditions of PDE3 inhibition, it further reduces sinoatrial rate. Both PDE3 and PDE4 control atrial and ventricular positive inotropic effects of (-)-noradrenaline. In contrast, neither PDE3 nor PDE4 limit the sinoatrial tachycardia induced by (-)-noradrenaline. In the rat, only PDE4, but not PDE1, PDE2, PDE3 and PDE5, reduces basal sinoatrial rate. None of the five rat PDEs limits the (-)-noradrenaline-evoked tachycardia. Taken together, these results confirm and expand evidence for our proposal that the cAMP-compartment modulating basal sinoatrial rate, controlled by PDE3 and/or PDE4, is different from the PDE-resistant cAMP compartment involved in beta(1)-adrenoceptor-mediated sinoatrial tachycardia.


Subject(s)
Cyclic AMP/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Adrenergic, beta-1/metabolism , Tachycardia/physiopathology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Disease Models, Animal , Female , Male , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/drug effects , Rabbits , Rats , Rats, Sprague-Dawley , Sinoatrial Node/physiopathology
16.
Br J Pharmacol ; 156(1): 62-83, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19133992

ABSTRACT

BACKGROUND AND PURPOSE: beta(1)- and beta(2)-adrenoceptors coexist in rat heart but beta(2)-adrenoceptor-mediated inotropic effects are hardly detectable, possibly due to phosphodiesterase (PDE) activity. We investigated the influence of the PDE3 inhibitor cilostamide (300 nmol x L(-1)) and the PDE4 inhibitor rolipram (1 micromol x L(-1)) on the effects of (-)-catecholamines. EXPERIMENTAL APPROACH: Cardiostimulation evoked by (-)-noradrenaline (ICI118551 present) and (-)-adrenaline (CGP20712A present) through beta(1)- and beta(2)-adrenoceptors, respectively, was compared on sinoatrial beating rate, left atrial and ventricular contractile force in isolated tissues from Wistar rats. L-type Ca(2+)-current (I(Ca-L)) was assessed with whole-cell patch clamp. KEY RESULTS: Rolipram caused sinoatrial tachycardia. Cilostamide and rolipram did not enhance chronotropic potencies of (-)-noradrenaline and (-)-adrenaline. Rolipram but not cilostamide potentiated atrial and ventricular inotropic effects of (-)-noradrenaline. Cilostamide potentiated the ventricular effects of (-)-adrenaline but not of (-)-noradrenaline. Concurrent cilostamide + rolipram uncovered left atrial effects of (-)-adrenaline. Both rolipram and cilostamide augmented the (-)-noradrenaline (1 micromol x L(-1)) evoked increase in I(Ca-L). (-)-Adrenaline (10 micromol x L(-1)) increased I(Ca-L) only in the presence of cilostamide but not rolipram. CONCLUSIONS AND IMPLICATIONS: PDE4 blunts the beta(1)-adrenoceptor-mediated inotropic effects. PDE4 reduces basal sinoatrial rate in a compartment distinct from compartments controlled by beta(1)- and beta(2)-adrenoceptors. PDE3 and PDE4 jointly prevent left atrial beta(2)-adrenoceptor-mediated inotropy. Both PDE3 and PDE4 reduce I(Ca-L) responses through beta(1)-adrenoceptors but the PDE3 component is unrelated to inotropy. PDE3 blunts both ventricular inotropic and I(Ca-L) responses through beta(2)-adrenoceptors.


Subject(s)
Calcium Channels, L-Type/physiology , Heart Rate/drug effects , Myocardial Contraction/drug effects , Phosphodiesterase 3 Inhibitors , Phosphodiesterase 4 Inhibitors , Receptors, Adrenergic, beta-1/physiology , Receptors, Adrenergic, beta-2/physiology , Animals , Atrial Function/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 3/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Dose-Response Relationship, Drug , Epinephrine/pharmacology , In Vitro Techniques , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Norepinephrine/pharmacology , Patch-Clamp Techniques , Quinolones/pharmacology , Rats , Rats, Wistar , Rolipram/pharmacology , Ventricular Function/drug effects
17.
Br J Pharmacol ; 156(2): 237-49, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19154438

ABSTRACT

BACKGROUND AND PURPOSE: Atrial inotropic responses to 5-HT mediated through 5-HT(4) receptors fade, presumably through phosphodiesterase (PDE) activity. We investigated the influence of a selective inhibitor of PDE3 (cilostamide) or of PDE4 (rolipram) on the fade of 5-HT responses in atrial muscle. EXPERIMENTAL APPROACH: 5-HT responses were compared, ex vivo, on sinoatrial beating rate of newborn piglets, porcine atrial and ventricular force, and human atrial force. cAMP levels were assessed in piglet atrium. KEY RESULTS: 5-HT-evoked sinoatrial tachycardia did not fade and was not potentiated by cilostamide (300 nmol.L(-1)) or rolipram (1 micromol.L(-1)). Inotropic responses to 5-HT faded in atria from piglets, adolescent pigs and humans. Cilostamide reduced atrial fade of 5-HT responses in adolescent pigs and humans but not in newborn piglets. Cilostamide disclosed 5-HT ventricular responses in newborn, but not adolescent pigs. Rolipram reduced fade of atrial 5-HT responses in newborn and adolescent pigs but not in humans. Concurrent cilostamide + rolipram abolished fade of 5-HT responses in porcine left atria and facilitated ventricular 5-HT responses, but did not reduce residual fade in human atrium in the presence of cilostamide. 5-HT-evoked increases in cAMP faded; fade was abolished by concurrent cilostamide + rolipram. CONCLUSIONS AND IMPLICATIONS: PDE3-induced control of porcine 5-HT responses differed in atrium and ventricle and changed with age. PDE3 and PDE4 jointly prevented fade of inotropic and cAMP responses to 5-HT in porcine atrium. Unlike porcine atria, only PDE3 induced fade of 5-HT responses in human atria.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Heart Rate/drug effects , Muscle Contraction/drug effects , Receptors, Serotonin, 5-HT4/physiology , Age Factors , Animals , Animals, Newborn , Arrhythmia, Sinus/physiopathology , Atrial Function/drug effects , Calcium Channels, L-Type/physiology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Female , Humans , In Vitro Techniques , Male , Middle Aged , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Phosphodiesterase 3 Inhibitors , Phosphodiesterase 4 Inhibitors , Quinolones/pharmacology , Rolipram/pharmacology , Serotonin/pharmacology , Species Specificity , Swine , Ventricular Function/drug effects
18.
J Pharmacol Exp Ther ; 328(1): 213-22, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18820136

ABSTRACT

(-)-Isoproterenol [4-[1-hydroxy-2-[(1-methylethyl)amino]ethyl]-1,2-benzene diol hydrochloride] relaxes murine detrusor through beta-adrenoceptors (ARs); however, the beta-AR subtypes involved are unknown. beta(2)-ARs have been associated with caveolae, plasma-lemmal scaffolding domains that are absent in caveolin-1 (cav-1) knockout (KO) mice. Here, we studied detrusor responses in the absence and presence of beta-AR subtype-selective antagonists in wild-type (WT) and cav-1 KO mice. To inquire whether the murine detrusor model is relevant to man, beta-AR subtypes that mediate (-)-isoproterenol-evoked human detrusor relaxation were investigated. In WT mice, (-)-isoproterenol concentration-dependently relaxed the KCl (40 mM)-precontracted detrusor (-logEC(50)M = 8.04, E(max) = 62%). The effects of (-)-isoproterenol were surmountably antagonized by the beta(2)-AR-selective antagonist ICI 118,551 [(+/-)-1-[2,3-(dihydro-7-methyl-1H-inden-4-yl)oxy]-3-[(1-methylethyl)amino]-2-butanol] (pK(B) = 9.28) but not affected by the beta(1)-AR-selective antagonist CGP 20712 [1-[2-((3-carbamoyl-4-hydroxy)phenoxy)ethylamino]-3-[4-(1-methyl-4-trifluoromethyl-2-imidazolyl)phenoxy]-2-propanol] and beta(3)-AR-selective L-748,337 [(S)-M-[4-[2-[3-[3-[acetamidomethyl)phenoxy)-2-hydroxypropyl]-amino]-ethyl]-phenylbenzsulfonamide)], suggesting involvement of beta(2)-AR only. The cav-1 KO detrusor displayed significant contractile dysfunction. (-)-Isoproterenol was less potent and efficient in relaxing detrusor from cav-1 KO (-logEC(50)M, 7.76; E(max) = 44%), but ICI 118,551 caused similar antagonism (pK(B) = 9.15), suggesting that beta(2)-AR function persisted in cav-1 KO. The beta(3)-AR-selective antagonist L-748,337 in the presence of ICI 118,551 and CGP 20712 caused additional blockade of (-)-isoproterenol effects in cav-1 KO, consistent with a beta(3)-AR involvement during relaxation and suppression of this effect in WT. (-)-Isoproterenol relaxed human detrusor muscle precontracted with carbachol (-logEC(50)M = 6.39, E(max) = 52%). However, the effects of (-)-isoproterenol in human detrusor were not blocked by CGP 20712 or ICI 118,551 but antagonized by L-748,337 (pK(B) = 7.65). We conclude that murine detrusor relaxation occurs via beta(2)-AR, and loss of caveolae does not perturb beta(2)-AR function but unmasks an additional activation of beta(3)-AR. In contrast, detrusor relaxation in man is mediated exclusively via beta(3)-AR.


Subject(s)
Catecholamines/physiology , Muscle Relaxation/physiology , Muscle, Skeletal/physiology , Receptors, Adrenergic, beta-2/physiology , Receptors, Adrenergic, beta-3/physiology , Urinary Bladder/physiology , Aged , Animals , Caveolin 1/deficiency , Caveolin 1/genetics , Caveolin 1/physiology , DNA Primers , Epinephrine/pharmacology , Female , Humans , Isoproterenol/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mucous Membrane/physiology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Relaxation/drug effects , Polymerase Chain Reaction , RNA, Messenger/genetics , Receptors, Adrenergic, beta-2/genetics
19.
Naunyn Schmiedebergs Arch Pharmacol ; 379(4): 379-84, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19005642

ABSTRACT

Acting through a low-affinity site of the beta(1)-adrenoceptor (beta(1L)AR), CGP12177 causes sinoatrial tachycardia and positive inotropic effects in left atrium but not in the ventricle of the rat. However, inhibition of either PDE3 or PDE4 also uncovers positive inotropic effects of CGP12177 in ventricle, but whether these phosphodiesterases also control the atrial agonist effects of CGP12177 was unknown. We, therefore, investigated the effects of the PDE3-selective inhibitor cilostamide (300 nM) and PDE4 inhibitor rolipram (1 microM) on the (-)-CGP12177-evoked increases of sinoatrial beating rate and force of paced left atria of the rat. Rolipram (n = 8) increased basal sinoatrial rate by 27 +/- 5 bpm but cilostamide (n = 8) had no effect. The chronotropic potency of (-)-CGP12177 (-logEC(50)M = 7.5) was not changed by rolipram and cilostamide or their combination. (-)-CGP12177 increased left atrial force with intrinsic activity 0.25 compared to (-)-isoprenaline. Rolipram (n = 8) and cilostamide (n = 8) did not change basal force of left atria but concurrent rolipram + cilostamide (n = 8) increased force by 52 +/- 9% of the effect of 200 microM (-)-isoprenaline. Neither rolipram nor cilostamide affected the inotropic potency of (-)-CGP12177 (-logEC(50)M = 7.4) but concurrent rolipram + cilostamide caused potentiation (-logEC(50)M = 8.2) and converted (-)-CGP12177 into a full agonist compared to (-)-isoprenaline. Cyclic AMP appears to maintain sinoatrial rate and PDE4 elicits bradycardia through hydrolysis of cAMP in a compartment distinct from the beta(1L)AR-induced cAMP compartment through which (-)-CGP12177 causes tachycardia. In contrast to the (-)-CGP12177-evoked tachycardia, not controlled by PDE3 and PDE4, these isoenzymes jointly reduce (-)-CGP12177-evoked increases of left atrial contractility through beta(1L)AR.


Subject(s)
Arrhythmia, Sinus/chemically induced , Cyclic Nucleotide Phosphodiesterases, Type 3/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Heart Atria/drug effects , Heart Rate/physiology , Myocardial Contraction/physiology , Propanolamines/pharmacology , Adrenergic beta-1 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Animals , Arrhythmia, Sinus/physiopathology , Atrial Function, Left/drug effects , Atrial Function, Left/physiology , Atrial Function, Right/drug effects , Atrial Function, Right/physiology , Bradycardia/metabolism , Drug Partial Agonism , Female , Heart Atria/physiopathology , Heart Rate/drug effects , In Vitro Techniques , Isoproterenol/pharmacology , Male , Myocardial Contraction/drug effects , Phosphodiesterase 3 Inhibitors , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Quinolones/pharmacology , Rats , Rats, Sprague-Dawley , Rolipram/pharmacology , Stimulation, Chemical , Tachycardia/chemically induced , Tachycardia/physiopathology
20.
Pharmacol Ther ; 118(3): 303-36, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18501968

ABSTRACT

beta-Adrenoceptor blocking agents (beta-blockers) that at low concentrations antagonize cardiostimulant effects of catecholamines, but at high concentrations also cause cardiostimulation, have been appearing since the late 1960s. These cardiostimulant beta-blockers, coined non-conventional partial agonists, antagonize the effects of catecholamines through a high-affinity site (beta(1H)AR), but cause cardiostimulation mainly through a low-affinity site (beta(1L)AR) of the myocardial beta(1)-adrenoceptor. The experimental non-conventional partial agonist (-)-CGP12177 increases cardiac L-type Ca(2+) current density and Ca(2+) transients, shortens action potential duration but augments action potential plateau, increases heart rate and force, as well as causes arrhythmic Ca(2+) transients and arrhythmic cardiocyte contractions. Other beta-blockers, which do not cause cardiostimulation, consistently have lower affinity for beta(1L)AR than beta(1H)AR. These sites were verified and the cardiac pharmacology of non-conventional partial agonists confirmed on recombinant beta(1)-adrenoceptors and on beta(1)-adrenoceptors overexpressed into the heart. A targeted mutation of Asp138 to Glu138 virtually abolished the pharmacology of beta(1H)AR but left intact the pharmacology of beta(1L)AR. Non-conventional partial agonists may be beneficial for the treatment of peripheral autonomic neuropathy but probably due to their arrhythmic propensities, may be harmful for the treatment of chronic heart failure.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Receptors, Adrenergic, beta-1/drug effects , Adrenergic beta-Agonists/adverse effects , Adrenergic beta-Agonists/history , Adrenergic beta-Antagonists/adverse effects , Adrenergic beta-Antagonists/history , Animals , Binding Sites , Catecholamines/antagonists & inhibitors , Heart Failure/drug therapy , Heart Failure/physiopathology , History, 20th Century , Humans , Receptors, Adrenergic, beta-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...