Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Andrology ; 2(6): 955-66, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25270366

ABSTRACT

The use of clonidine, a selective agonist of α2-adrenoceptors, is related to the fertility impairment. Thus, it has been described that changes in the epididymal function are related to the loss of fertility. Therefore, this study was sought to further evaluate the effects of clonidine in the rat distal cauda epididymis contractions and its consequence in the sperm parameters. The in vitro effects of clonidine in the isolated distal cauda epididymis were evaluated by pharmacological experiments. The consecutive contractile responses for clonidine in distal cauda epididymis showed desensitization. The noradrenaline-induced contractions were desensitized after in vitro clonidine pre-treatment (10(-5) M for 10 min). Clonidine was unable to alter the noradrenaline contractions if the in vitro pre-treatment was made in the presence of idazoxan (α2-adrenoceptor antagonist), whereas prazosin (α1-adrenoceptor antagonist) was ineffective. Moreover, the in vitro clonidine pre-treatment increased frequency and amplitude of spontaneous contraction of distal cauda epididymis. In addition, to induce in vivo desensitization of α2-adrenoceptors, male Wistar rats were treated with crescent doses of clonidine and distal cauda of epididymis contraction and sperm parameters were analyzed. The in vivo treatment with clonidine diminished the potency of the contractions induced by adrenergic agonists and augmented the frequency and amplitude of spontaneous contraction of distal cauda epididymis. This treatment also altered the sperm transit time in epididymis, epididymal sperm reserves, sperm lipid peroxidation, and antioxidant enzymes activity. The results suggest that clonidine was able to affect the sperm quantity and quality by decreasing the transit time related to the increase in the frequency and amplitude of spontaneous contractions in epididymis, although the contractions induced by adrenergic agonists were desensitized.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Clonidine/pharmacology , Epididymis/drug effects , Spermatozoa/drug effects , Animals , Epididymis/physiology , Male , Muscle Contraction/drug effects , Rats , Rats, Wistar
2.
Pflugers Arch ; 442(3): 376-82, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11484768

ABSTRACT

Carbachol-induced contractions of rat stomach fundus strips, obtained in a nutrient solution containing 1.8 mM Ca2+, were resistant to Ca2+ withdrawal, even after 1 h of bathing the tissues in a nominal 0 Ca2+ solution. This was not observed when K+ was used to evoke contractions, which were rapidly inhibited after Ca2+ removal (t1/2=2 min). The effect of carbachol in 0 Ca2+ solution was reduced by using drugs that reduce intracellular pools of Ca2+, such as caffeine (1-3 mM), ryanodine (30 microM) or thapsigargin (1 microM), corroborating the involvement of intracellular Ca2+ stores. On the other hand, when the 0 Ca2+ solution contained EGTA, a complete decline of carbachol effects was observed within about 8 min, indicating the involvement of extracellular Ca2+. Atomic absorption spectrometry showed that our 0 Ca2+ solution still contained 45 microM Ca2+, which was drastically reduced to 5.9 nM in the presence of EGTA. Taken together, our results indicate that the effects of carbachol are due to the mobilization of caffeine-, ryanodine- and thapsigargin-sensitive intracellular Ca2+ stores, and that these stores are not inactivated or depleted if micromolar concentrations (45 microM), but not nanomolar concentrations (5.9 nM) of Ca2+ are maintained in the extracellular milieu.


Subject(s)
Calcium/metabolism , Gastric Fundus/physiology , Receptors, Muscarinic/metabolism , Animals , Biological Transport/drug effects , Caffeine/pharmacology , Calcium/pharmacology , Carbachol/pharmacology , Chelating Agents/pharmacology , Cholinergic Agonists/pharmacology , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Female , Muscle, Smooth/physiology , Nickel/pharmacology , Peristalsis/drug effects , Phosphodiesterase Inhibitors/pharmacology , Rats , Rats, Inbred WF , Ryanodine/pharmacology , Thapsigargin/pharmacology
3.
Br J Pharmacol ; 127(8): 1832-6, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10482913

ABSTRACT

1. The actions of the alpha1-adrenoceptor antagonist indoramin have been examined against the contractions induced by noradrenaline in the rat vas deferens and aorta taking into account a putative neuronal uptake blocking activity of this antagonist which could result in self-cancelling actions. 2. Indoramin behaved as a simple competitive antagonist of the contractions induced by noradrenaline in the vas deferens and aorta yielding pA2 values of 7.38+/-0.05 (slope=0.98+/-0.03) and 6.78+/-0.14 (slope=1.08+/-0.06), respectively. 3. When the experiments were repeated in the presence of cocaine (6 microM) the potency (pA2) of indoramin in antagonizing the contractions of the vas deferens to noradrenaline was increased to 8.72+/-0.07 (slope=1.10+/-0.05) while its potency remained unchanged in the aorta (pA2=6.69+/-0.12; slope=1.04+/-0.05). 4. In denervated vas deferens, indoramin antagonized the contractions to noradrenaline with a potency similar to that found in the presence of cocaine (8.79+/-0.07; slope=1.09+/-0.06). 5. It is suggested that indoramin blocks alpha1-adrenoceptors and neuronal uptake in rat vas deferens resulting in Schild plots with slopes not different from unity even in the absence of selective inhibition of neuronal uptake. As a major consequence of this double mechanism of action, the pA2 values for this antagonist are underestimated when calculated in situations where the neuronal uptake is active, yielding spurious pKB values.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-Antagonists/pharmacology , Aorta/drug effects , Indoramin/pharmacology , Vas Deferens/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Cocaine/pharmacology , Male , Norepinephrine/antagonists & inhibitors , Norepinephrine/pharmacology , Rats , Rats, Wistar
4.
Biochim Biophys Acta ; 1419(2): 343-52, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10407085

ABSTRACT

The inward currents in single smooth muscle cells (SMC) isolated from epididymal part of rat vas deferens have been studied using whole-cell patch-clamp method. Depolarising steps from holding potential -90 mV evoked inward current with fast and slow components. The component with slow activation possessed voltage-dependent and pharmacological properties characteristic for Ca(2+) current carried through L-type calcium channels (I(Ca)). The fast component of inward current was activated at around -40 mV, reached its peak at 0 mV, and disappeared upon removal of Na ions from bath solution. This current was blocked in dose-dependent manner by tetrodotoxin (TTX) with an apparent dissociation constant of 6.7 nM. On the basis of voltage-dependent characteristics, TTX sensitivity of fast component of inward current and its disappearance in Na-free solution it is suggested that this current is TTX-sensitive depolarisation activated sodium current (I(Na)). Cell dialysis with a pipette solution containing no macroergic compounds resulted in significant inhibition of I(Ca) (depression of peak I(Ca) by about 81% was observed by 13 min of dialysis), while I(Na) remained unaffected during 50 min of dialysis. These data draw first evidence for the existence of TTX-sensitive Na(+) current in single SMC isolated from rat vas deferens. These Na(+) channels do not appear to be regulated by a phosphorylation process under resting conditions.


Subject(s)
Calcium Channels/drug effects , Muscle, Smooth/metabolism , Nifedipine/pharmacology , Sodium Channels/drug effects , Tetrodotoxin/pharmacology , Vas Deferens/metabolism , Animals , Calcium Channels/metabolism , Cell Separation , Dose-Response Relationship, Drug , Male , Membrane Potentials , Muscle, Smooth/cytology , Patch-Clamp Techniques , Phosphorylation , Rats , Rats, Wistar , Sodium Channels/metabolism , Vas Deferens/cytology
5.
Article in English | MEDLINE | ID: mdl-10797863

ABSTRACT

The contractile effect of serotonin was studied in rat vas deferens, in comparison with that of noradrenaline and tyramine, after reserpine treatment, surgical denervation, and transplantation to the colon. In reserpinized animals the effect of 5HT resembled that of tyramine, since it was strikingly reduced, in spite of a small residual effect, showing that in normal preparations the effects of 5HT and tyramine are predominantly due to the release of endogenous noradrenaline. However, in denervated or transplanted vas deferens, in which the effect of tyramine is also abolished, the effect of 5HT was potentiated. It is suggested that after chronic, long lasting depletion of endogenous noradrenaline, there are alternate mechanisms that are generated to improve the contractile effect of 5HT, but not of tyramine. The nature of these mechanisms is still unknown.


Subject(s)
Muscle Contraction/drug effects , Serotonin/pharmacology , Vas Deferens/drug effects , Adrenergic Uptake Inhibitors/pharmacology , Animals , Denervation , Dose-Response Relationship, Drug , Male , Norepinephrine/pharmacology , Rats , Rats, Wistar , Reserpine/pharmacology , Sympathomimetics/pharmacology , Tyramine/pharmacology , Vas Deferens/injuries , Vas Deferens/physiology
6.
Article in English | BINACIS | ID: bin-40115

ABSTRACT

The contractile effect of serotonin was studied in rat vas deferens, in comparison with that of noradrenaline and tyramine, after reserpine treatment, surgical denervation, and transplantation to the colon. In reserpinized animals the effect of 5HT resembled that of tyramine, since it was strikingly reduced, in spite of a small residual effect, showing that in normal preparations the effects of 5HT and tyramine are predominantly due to the release of endogenous noradrenaline. However, in denervated or transplanted vas deferens, in which the effect of tyramine is also abolished, the effect of 5HT was potentiated. It is suggested that after chronic, long lasting depletion of endogenous noradrenaline, there are alternate mechanisms that are generated to improve the contractile effect of 5HT, but not of tyramine. The nature of these mechanisms is still unknown.

7.
Mol Pharmacol ; 54(1): 220-30, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9658209

ABSTRACT

The slowly activating delayed rectifier K+ current, IKs, is an important modulator of cardiac action potential repolarization. Here, we describe a novel benzodiazepine, [L-364,373 [(3-R)-1, 3-dihydro-5-(2-fluorophenyl)-3-(1H-indol-3-ylmethyl)-1-methyl-2H- 1,4-benzodiazepin-2-one] (R-L3), that activates IKs and shortens action potentials in guinea pig cardiac myocytes. These effects were additive to isoproterenol, indicating that channel activation by R-L3 was independent of beta-adrenergic receptor stimulation. The increase of IKs by R-L3 was stereospecific; the S-enantiomer, S-L3, blocked IKs at all concentrations examined. The increase in IKs by R-L3 was greatest at voltages near the threshold for normal channel activation, caused by a shift in the voltage dependence of IKs activation. R-L3 slowed the rate of IKs deactivation and shifted the half-point of the isochronal (7.5 sec) activation curve for IKs by -16 mV at 0.1 microM and -24 mV at 1 microM. R-L3 had similar effects on cloned KvLQT1 channels expressed in Xenopus laevis oocytes but did not affect channels formed by coassembly of KvLQT1 and hminK subunits. These findings indicate that the association of minK with KvLQT1 interferes with the binding of R-L3 or prevents its action once bound to KvLQT1 subunits.


Subject(s)
Benzodiazepines/pharmacology , Heart/drug effects , Potassium Channels, Voltage-Gated , Potassium Channels/drug effects , Action Potentials/drug effects , Animals , Delayed Rectifier Potassium Channels , Guinea Pigs , Heart/physiology , Heart Ventricles/drug effects , Humans , Ion Channels/drug effects , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Potassium Channels/metabolism , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/metabolism , Ventricular Function , Xenopus laevis/genetics
8.
Br J Pharmacol ; 123(1): 23-30, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9484850

ABSTRACT

1. The class III antiarrhythmic azimilide has previously been shown to inhibit I(Ks) and I(Kr) in guinea-pig cardiac myocytes and I(Ks) (minK) channels expressed in Xenopus oocytes. Because HERG channels underly the conductance I(Kr), in human heart, the effects of azimilide on HERG channels expressed in Xenopus oocytes were the focus of the present study. 2. In contrast to other well characterized HERG channel blockers, azimilide blockade was reverse use-dependent, i.e., the relative block and apparent affinity of azimilide decreased with an increase in channel activation frequency. Azimilide blocked HERG channels at 0.1 and 1 Hz with IC50s of 1.4 microM and 5.2 microM respectively. 3. In an envelope of tail test, HERG channel blockade increased with increasing channel activation, indicating binding of azimilide to open channels. 4. Azimilide blockade of HERG channels expressed in Xenopus oocytes and I(Kr) in mouse AT-1 cells was decreased under conditions of high [K+]e, whereas block of slowly activating I(Ks) channels was not affected by changes in [K+]e. 5. In summary, azimilide is a blocker of cardiac delayed rectifier channels, I(Ks) and HERG. Because of the distinct effects of stimulation frequency and [K+]e on azimilide block of I(Kr) and I(Ks) channels, we conclude that the relative contribution of block of each of these cardiac delayed rectifier channels depends on heart frequency. [K+]e and regulatory status of the respective channels.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Cation Transport Proteins , DNA-Binding Proteins , Imidazoles/pharmacology , Imidazolidines , Piperazines/pharmacology , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Trans-Activators , Animals , Cell Line , ERG1 Potassium Channel , Electric Stimulation , Ether-A-Go-Go Potassium Channels , Guinea Pigs , Hydantoins , Membrane Potentials/physiology , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Potassium/pharmacology , Potassium Channels/metabolism , RNA, Messenger/biosynthesis , Xenopus
12.
Circulation ; 94(11): 2938-46, 1996 Dec 01.
Article in English | MEDLINE | ID: mdl-8941124

ABSTRACT

BACKGROUND: The class III antiarrhythmic agent RP 58866 and its active enantiomer, terikalant, are reported to selectively block the inward rectifier K+ current, IK1. These drugs have demonstrated efficacy in animal models of cardiac arrhythmias, suggesting that block of IK1 may be a useful antiarrhythmic mechanism. The symmetrical action potential (AP)-prolonging and bradycardic effects of these drugs, however, are inconsistent with a sole effect on IK1. METHODS AND RESULTS: We studied the effects of RP 58866 and terikalant on AP and outward K+ currents in guinea pig ventricular myocytes. RP 58866 and terikalant potently blocked the rapidly activating delayed rectifier K+ current, IKr, with IC50S of 22 and 31 nmol/L, respectively. Block of IK1 was approximately 250-fold less potent; IC50S were 8 and 6 mumol/L, respectively. No significant block of the slowly activating delayed rectifier, IK1, was observed at < or = 10 mumol/L. The phenotypical IKr currents in mouse AT-1 cells and Xenopus oocytes expressing HERG were also blocked 50% by 200 to 250 nmol/L RP 58866 or terikalant, providing further conclusive evidence for potent block of IKr. RP 58866 < or = 1 mumol/L and dofetilide increased AP duration symmetrically, consistent with selective block of IKr. Only higher concentrations (> or = 10 mumol/L) of RP 58866 slowed the rate of AP repolarization and decreased resting membrane potential, consistent with an additional but substantially less potent block of IK1. CONCLUSIONS: These data demonstrate that RP 58866 and terikalant are potent blockers of IKr and prompt a reinterpretation of previous studies that assumed specific block of IK1 by these drugs.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Cation Transport Proteins , Chromans/pharmacology , DNA-Binding Proteins , Heart/drug effects , Heart/physiology , Piperidines/pharmacology , Potassium Channels, Voltage-Gated , Potassium/antagonists & inhibitors , Trans-Activators , Action Potentials , Animals , ERG1 Potassium Channel , Electric Conductivity , Ether-A-Go-Go Potassium Channels , Guinea Pigs , Mice , Myocardium/cytology , Oocytes/metabolism , Osmolar Concentration , Patch-Clamp Techniques , Potassium/physiology , Potassium Channels/physiology , Reaction Time/drug effects , Stereoisomerism , Xenopus
13.
Am J Physiol ; 271(6 Pt 2): H2477-89, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8997308

ABSTRACT

The delayed rectifier K+ current (IK) in rabbit heart has long been thought to consist of only a single, rapidly activating, dofetilide-sensitive current, IKr. However, we find that IK of rabbit ventricular myocytes actually consists of both rapid and slow components, IKr and IKs, respectively, that can be isolated pharmacologically. Thus, after complete blockade of IKr with dofetilide, the remaining current, IKs, is homogeneous as judged by an envelope of tails test. IKs activates and deactivates slowly, continues to activate during sustained depolarizations, has a half-activation potential of 7.0 +/- 0.8 mV and slope factor of 11.0 +/- 0.7 mV, reverses at -77.2 +/- 1.3 mV (extracellular K+ concentration = 4 mM), is increased by removing extracellular K+, and is enhanced by isoproterenol and stocked by azimilide. Northern analysis demonstrates that the minK (IsK) gene, which encodes a subunit of the channel that underlies the IKs current, is expressed in rabbit heart. Expression of the rabbit protein in Xenopus oocytes elicits a slowly activating, voltage-dependent current, IsK, similar to those expressed previously from mouse, rat, guinea pig, and human genes. The results demonstrate that IKs is present in rabbit ventricle and therefore contributes to cardiac repolarization in this species.


Subject(s)
Imidazolidines , Potassium Channels/physiology , Ventricular Function , Adrenergic beta-Agonists/pharmacology , Amino Acid Sequence , Animals , Anti-Arrhythmia Agents/pharmacology , Electric Conductivity , Electrophysiology , Gene Expression , Guinea Pigs , Heart/physiology , Humans , Hydantoins , Imidazoles/pharmacology , Isoproterenol/pharmacology , Mice , Molecular Sequence Data , Mutation , Oocytes/metabolism , Piperazines/pharmacology , Potassium Channels/genetics , Rabbits , Rats , Xenopus laevis
14.
Eur J Pharmacol ; 307(3): 299-304, 1996 Jul 04.
Article in English | MEDLINE | ID: mdl-8836618

ABSTRACT

Experiments were performed with rat vas deferens to verify whether agmatine, an endogenous ligand for adrenoceptors and imidazoline receptors, can influence sympathetic neurotransmission, with respect to contractions induced by transmural nerve stimulation, contractions induced by exogenous noradrenaline, and overflow of endogenous noradrenaline. It was shown that agmatine (a) caused a dose-dependent potentiation of electrically induced twitches, up to about 70% in relation to controls, (b) shifted to the right the inhibitory concentration-response curves for clonidine on electrically induced twitches, indicating competitive antagonism at presynaptic alpha-adrenoceptors, with a pA2 value of 4.12 +/- 0.10, (c) shifted to the right the concentration-response curves for noradrenaline-induced contractions, indicating competitive antagonism at postsynaptic alpha-adrenoceptors as well, with a pA2 value of 4.03 +/- 0.10, and (d) caused a dose-dependent increase of KCI-induced overflow of noradrenaline, up to about 90% in relation to controls. It is concluded that agmatine has multiple effects on sympathetic neurotransmission in rat vas deferens.


Subject(s)
Agmatine/pharmacology , Muscle Contraction/drug effects , Synaptic Transmission/drug effects , Vas Deferens/drug effects , Animals , Clonidine/pharmacology , Electric Stimulation , In Vitro Techniques , Male , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Norepinephrine/metabolism , Norepinephrine/pharmacology , Rats , Rats, Wistar , Receptors, Adrenergic, alpha/drug effects , Vas Deferens/metabolism
15.
J Cardiovasc Pharmacol ; 26(2): 259-71, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7475051

ABSTRACT

We studied the effects of NE-10064 (azimilide), a new antiarrhythmic agent reported to be a selective blocker of the slowly activating component of the delayed rectifier, IKs. In ferret papillary muscles, NE-10064 increased effective refractory period (ERP) and decreased isometric twitch tension in a concentration-dependent manner (0.3-30 microM). Increases in ERP showed reverse use-dependence, and were greater at 1 than at 3 Hz. In contrast, changes in tension were use dependent, with larger decreases observed at 3 than at 1 Hz. In guinea pig ventricular myocytes, NE-10064 (0.3-3 microM) significantly prolonged action potential duration (APD) at 1 Hz. At 3 Hz, NE-10064 (0.3-1 microM) increased APD only slightly, and at 10 microM decreased APD and the plateau potential. NE-10064 potently blocked the rapidly activating component of the delayed rectifier, IKr (IC50 0.4 microM), and inhibited IKs (IC50 3 microM) with nearly 10-fold less potency. NE-10064 (10 microM) did not block the inward rectifier potassium current (IKl). NE-10064 (10 microM) blocked the L-type calcium current (ICa) in a use-dependent manner; block was greater at 3 than at 1 Hz. We conclude that (a) NE-10064's block of potassium currents is relatively selective for IKr over IKs, (b) NE-10064 inhibits ICa in a use-dependent fashion, and (c) NE-10064's effects on ERP and tension in papillary muscle as well as APD and action potential plateau level in myocytes may be explained by its potassium and calcium channel blocking properties.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Calcium Channel Blockers/pharmacology , Heart/drug effects , Imidazoles/pharmacology , Imidazolidines , Piperazines/pharmacology , Potassium Channel Blockers , Action Potentials/drug effects , Animals , Calcium/metabolism , Dose-Response Relationship, Drug , Ferrets , Guinea Pigs , Heart/physiology , Hydantoins , In Vitro Techniques , Male , Refractory Period, Electrophysiological/drug effects
17.
Circ Res ; 76(1): 110-9, 1995 Jan.
Article in English | MEDLINE | ID: mdl-8001268

ABSTRACT

We compared the cardiac electrophysiological actions of two types of H1-receptor antagonists--the piperidines, astemizole and terfenadine, and the nonpiperidines, chlorpheniramine and pyrilamine-in vitro in guinea pig ventricular myocytes and in vivo in chloralose-anesthetized dogs. Astemizole and terfenadine significantly increased action potential duration of guinea pig myocytes. This concentration-dependent prolongation of action potential duration was reverse frequency dependent and led to development of early afterdepolarizations, which occurred more frequently at higher concentrations and slower pacing frequencies. Astemizole and terfenadine potently blocked the rapidly activating component of the delayed rectifier, IKr, with IC50 values of 1.5 and 50 nmol/L, respectively. At 10 mumol/L, terfenadine but not astemizole blocked the slowly activating component of the delayed rectifier, IKs (58.4 +/- 3.1%), and the inward rectifier, IK1 (20.5 +/- 3.4%). Chlorpheniramine and pyrilamine blocked IKr relatively weakly (IC50 = 1.6 and 1.1 mumol/L, respectively) and IKs and IK1 less than 20% at 10 mumol/L. Astemizole and terfenadine (1.0 to 3.0 mg/kg IV) significantly prolonged the QTc interval and ventricular effective refractory period in vivo. Chlorpheniramine and pyrilamine (< or = 3.0 mg/kg) did not significantly affect these parameters. Block of repolarizing K+ currents, particularly IK1, by astemizole and terfenadine produces reverse rate-dependent prolongation of action potential duration and development of early afterdepolarizations, delays ventricular repolarization, and may underlie the development of torsade de pointes ventricular arrhythmias observed with the use and abuse of these agents.


Subject(s)
Astemizole/pharmacology , Chlorpheniramine/pharmacology , Heart/drug effects , Pyrilamine/pharmacology , Terfenadine/pharmacology , Action Potentials/drug effects , Animals , Dose-Response Relationship, Drug , Electrocardiography , Guinea Pigs , Heart/physiology , In Vitro Techniques , Potassium Channels/drug effects , Refractory Period, Electrophysiological/drug effects
18.
Eur J Pharmacol ; 256(3): 329-33, 1994 May 02.
Article in English | MEDLINE | ID: mdl-8045278

ABSTRACT

Radioligand binding assays were performed with the selective antagonist of dihydropyridine-sensitive Ca2+ channels [3H]PN200-110 (isradipine) in rat vas deferens, before and 7 days after denervation, and data were compared with those obtained for K(+)-induced contractions, which are Ca(2+)-dependent. The density (Bmax) of dihydropyridine binding sites was decreased to almost one-third of its normal value after denervation. The respective affinity (KD) was not significantly changed. In addition, it was observed that the K(+)-induced tonic contraction, which corresponded to 55 +/- 2% of the respective phasic contraction, was decreased to 41 +/- 3% after denervation. It is assumed that the decreased density of Ca2+ channels causes a decrease in K(+)-induced influx of Ca2+ and consequently of the corresponding tonic contraction. These results indicate that autonomic innervation can regulate the density of dihydropyridine-sensitive Ca2+ channels in the rat vas deferens.


Subject(s)
Isradipine/metabolism , Vas Deferens/metabolism , Animals , Binding Sites/drug effects , Calcium Channels/drug effects , Denervation , Dihydropyridines/pharmacology , Male , Muscle Contraction/drug effects , Potassium Chloride/pharmacology , Rats , Rats, Wistar , Vas Deferens/drug effects
19.
J Pharmacol Exp Ther ; 269(2): 541-54, 1994 May.
Article in English | MEDLINE | ID: mdl-7802864

ABSTRACT

The cardiac electrophysiologic and antiarrhythmic actions of two Class III ketone- and alcohol-containing spirobenzopyran piperidine analogs, L-702,958 and L-706,000 [MK-499], respectively, were assessed in vitro and in vivo. L-702,958 and L-706,000 [MK-499] selectively blocked the rapidly activating component of the delayed rectifier K+ current in guinea pig isolated ventricular myocytes (IC50 values, 14.6 and 43.9 nM, respectively), and prolonged effective refractory period in ferret isolated papillary muscles (EC25 values, 10.5 and 53.8 nM, respectively). In anesthetized dogs, L-702,958 and L-706,000 [MK-499] increased ventricular refractory periods (ED20 values, 3.3 and 9.2 micrograms/kg i.v., respectively) and concomitantly increased ECG QT interval and left ventricular+dP/dt. Cumulative i.v. administrations of up to 100 micrograms/kg of L-702,958 and 300 micrograms/kg L-706,000 [MK-499] in anesthetized dogs increased atrial and ventricular refractoriness and prolonged the ECG QT interval, but did not alter atrial, atrioventricular nodal, His-Purkinje or ventricular conduction indices. In anesthetized dogs studied chronically (9.2 +/- 1.1 days) after anterior myocardial infarction, the cumulative i.v. administrations of 100 micrograms/kg of L-702,958 and 300 of micrograms/kg L-706,000 [MK-499] suppressed the induction of ventricular tachyarrhythmia by programmed ventricular stimulation (suppression rates: 8 of 10, 80% and 9 of 11, 82%, respectively) and reduced the incidence of lethal ventricular arrhythmias (incidence of lethal ischemic arrhythmias: 4 of 10, 40% and 1 of 11 9%, respectively, compared to 34 of 40, 85%, in vehicle controls. L-702,958 and L-706,000 [MK-499] (cumulative 100 and 300 micrograms/kg i.v., respectively) did not facilitate the induction of arrhythmias by programmed ventricular stimulation in postinfarction dogs. After equivalently effective p.o. doses in conscious dogs, L-702,958 (10 micrograms/kg) and L-706,000 [MK-499] (30 micrograms/kg) increased ECG QT interval with long durations of action of approximately 9 and 14 hr, respectively. L-706,000 [MK-499] elicited a more consistent and sustained prolongation of the QT interval than L-702,958. These findings show that both L-702,958 and L-706,000 [MK-499] are potentially useful agents for the prevention of malignant ventricular arrhythmias in the setting of myocardial ischemic injury.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Benzopyrans/pharmacology , Heart/drug effects , Piperidines/pharmacology , Animals , Dogs , Electrocardiography/drug effects , Female , Ferrets , Guinea Pigs , Heart/physiology , Hemodynamics/drug effects , In Vitro Techniques , Male , Molecular Structure , Myocardial Ischemia/physiopathology
20.
Proc Natl Acad Sci U S A ; 91(5): 1766-70, 1994 Mar 01.
Article in English | MEDLINE | ID: mdl-7510407

ABSTRACT

We have isolated cardiac cDNA and genomic clones encoding the guinea pig IsK protein. The deduced amino acid sequence is approximately 78% identical to the rat, mouse, and human variants of this channel, and the structure of the gene encoding the protein is also similar to that in other species. For example, the gene is present only once in the haploid genome, the protein-coding sequence is present on a single uninterrupted exon, an intron exists in the 5' untranslated domain, and multiple alternative polyadenylation sites are used in processing the transcript. Expression of the guinea pig protein in Xenopus oocytes results in a slowly activating, voltage-dependent K+ current, IsK, similar to those expressed previously from the rat, mouse, and human genes. However, in sharp contrast to the rat and mouse currents, activation of protein kinase C with phorbol esters increases the amplitude of the guinea pig IsK current, analogous to its effects on the endogenous IKs current in guinea pig cardiac myocytes. Mutagenesis of the guinea pig cDNA to alter four cytoplasmic amino acid residues alters the phenotype of the current response to protein kinase C from enhancement to inhibition, mimicking that of rat and mouse IsK currents. This mutation is consistent with reports that phosphorylation of Ser-102 by protein kinase C decreases the current amplitude. These data explain previously reported differences in the regulatory properties between recombinant rat or mouse IsK channels and native guinea pig IKs channels and provide further evidence that the IsK protein forms the channels that underlie the IKs current in the heart.


Subject(s)
Membrane Proteins/metabolism , Myocardium/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels , Potassium/metabolism , Protein Kinase C/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Enzyme Activation , Female , Guinea Pigs , Humans , Ion Channels/metabolism , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Oocytes/metabolism , Rats , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...