Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 150(1): 37-46, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17128289

ABSTRACT

BACKGROUND AND PURPOSE: Ivabradine, a specific and use-dependent I(f) inhibitor, exerts anti-ischaemic activity purely by reducing heart rate. The aim of this work was to characterize its effect on the predominant HCN channel isoform expressed in human sino-atrial nodes (hSAN), to determine its kinetics in HCN channels from multicellular preparations and rate-dependency of its action. EXPERIMENTAL APPROACH: RT-PCR analysis of the four HCN channel isoforms was carried out on RNAs from hSAN. Patch-clamp and intracellular recordings were obtained from CHO cells stably expressing hHCN4 and isolated SAN, respectively. Beating rate of rat isolated atria was followed using a transducer. KEY RESULTS: hHCN4 mRNAs were predominant in hSAN. Ivabradine induced a time-dependent inhibition of hHCN4 with an IC(50) of 0.5 microM. In rabbit SAN, ivabradine progressively reduced the frequency of action potentials: by 10% after 3 h at 0.1 microM, by 14% after 2 h at 0.3 microM and by 17% after 1.5 h at 1 microM. After 3h, ivabradine reduced the beating rate of rat right atria with an IC(30) of 0.2 microM. The onset of action of ivabradine was use-dependent rather than time-dependent with slower effects than caesium, an extracellular I (f) blocker. Ivabradine 3 microM decreased the frequency of action potentials in SAN from guinea-pig, rabbit and pig by 33%, 21% and 15% at 40 min, respectively. CONCLUSIONS AND IMPLICATIONS: The use-dependent inhibition of hHCN4 current by ivabradine probably contributes to its slow developing effect in isolated SAN and right atria and to its increased effectiveness in species with rapid SAN activity.


Subject(s)
Benzazepines/pharmacology , Biological Clocks/drug effects , Heart Rate/drug effects , Ion Channels/antagonists & inhibitors , Muscle Proteins/antagonists & inhibitors , Action Potentials/drug effects , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclic Nucleotide-Gated Cation Channels , Female , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/genetics , Ivabradine , Male , Muscle Proteins/genetics , Potassium Channels , RNA, Messenger/genetics , Rabbits , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Swine
2.
J Pharmacol Exp Ther ; 298(2): 565-80, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11454918

ABSTRACT

S33005 displayed marked affinity for native, rat, and cloned human serotonin (5-HT) transporters (SERT) and less pronounced affinity for norepinephrine (NE) transporters (NET), while its affinity at dopamine (DA) transporters and >50 other sites was negligible. Reuptake of 5-HT and (less potently) NE into cerebral synaptosomes was inhibited by S33005, whereas DA reuptake was little affected. In vivo, S33005 prevented depletion of cerebral pools of 5-HT by parachloroamphetamine. Furthermore, it decreased electrical activity of raphe-localized serotonergic neurones, an action abolished by the 5-HT1A antagonist WAY100,635. At higher doses, S33005 blocked firing of locus ceruleus-localized adrenergic neurones, an action abolished by the alpha2-adrenergic antagonist idazoxan. In contrast, S33005 did not inhibit ventrotegmental dopaminergic neurones. In frontal cortex of freely moving rats, S33005 dose dependently elevated dialysate levels of 5-HT, NE, and DA. In hippocampus, levels of 5-HT and NE were similarly elevated, while in nucleus accumbens and striatum, levels of 5-HT were increased whereas DA was unaffected. Upon chronic (2 weeks) administration, basal levels of NE were elevated in frontal cortex and, therein, 5-HT2A receptor density was decreased. Comparative studies with clinically used antidepressants showed that venlafaxine possessed a profile similar to S33005 but was less potent. Clomipramine likewise interacted with SERTs and NETs but also with several other receptors types, while citalopram and reboxetine were preferential ligands of SERTs and NETs, respectively. In conclusion, S33005 interacts potently with SERTs and, less markedly, with NETs. It enhances extracellular levels of 5-HT and NE throughout corticolimbic structures and selectively elevates dialysis levels of DA in frontal cortex versus subcortical regions.


Subject(s)
Carrier Proteins/metabolism , Cyclohexanols/pharmacology , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Agents/pharmacology , Symporters , Animals , Brain Chemistry/drug effects , Carrier Proteins/drug effects , Citalopram/pharmacology , Clomipramine/pharmacology , Electrophysiology , Humans , Ligands , Male , Membrane Glycoproteins/drug effects , Morpholines/pharmacology , Norepinephrine Plasma Membrane Transport Proteins , Rats , Rats, Wistar , Reboxetine , Receptors, Adrenergic, alpha/metabolism , Receptors, Serotonin/metabolism , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins , Venlafaxine Hydrochloride , p-Chloroamphetamine/antagonists & inhibitors , p-Chloroamphetamine/pharmacology
3.
J Pharmacol Exp Ther ; 298(2): 581-91, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11454919

ABSTRACT

Reflecting its potent inhibition of serotonin (5-HT) reuptake (accompanying paper), S33005 blocked spontaneous tail-flicks induced by parachloroamphetamine in rats. This action was mimicked by the 5-HT reuptake inhibitor, citalopram, and the 5-HT/norepinephrine (NE) reuptake inhibitor, venlafaxine, whereas the preferential NE reuptake inhibitor, reboxetine, was inactive. Consistent with its less potent interaction with NE transporters, higher doses of S33005 attenuated induction of hypothermia by reserpine, an action mimicked by reboxetine and venlafaxine, whereas citalopram was ineffective. In mice, S33005 reduced immobility in forced-swim and tail-suspension procedures. It also inhibited marble-burying behavior and suppressed aggressive behavior between resident and intruder animals. In rats, S33005 generalized to a discriminative stimulus elicited by citalopram and attenuated hypnotic-sedative actions of the alpha2-adrenoceptor agonist, S18616. For these parameters, S33005 was a more potent agent (median, 1.2 mg/kg, s.c.) than venlafaxine, citalopram, reboxetine, or the tricyclic agent, clomipramine. Even at markedly higher doses (40.0-80.0 mg/kg, s.c.), S33005 little affected motor behavior. S33005 (10.0 mg/kg, s.c.) also increased responses in a learned helplessness paradigm in rats, whereas venlafaxine was ineffective. Finally, in a rat chronic mild-stress model, S33005 dose- (2.5-40.0 mg/kg) and time- (2-5 weeks) dependently enhanced sucrose consumption. Venlafaxine was likewise active in this procedure. In conclusion, in line with its inhibition of 5-HT and (less potently) NE reuptake, S33005 is active in a broad range of models suggestive of antidepressant activity. It exerts its actions more potently than venlafaxine and clomipramine, and its overall profile is distinct from those of citalopram and reboxetine.


Subject(s)
Behavior, Animal/drug effects , Carrier Proteins/metabolism , Cyclohexanols/pharmacology , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Agents/pharmacology , Symporters , Animals , Antipsychotic Agents/pharmacology , Body Temperature/drug effects , Carrier Proteins/drug effects , Citalopram/pharmacology , Clomipramine/pharmacology , Discrimination, Psychological/drug effects , Male , Membrane Glycoproteins/drug effects , Mice , Morpholines/pharmacology , Norepinephrine Plasma Membrane Transport Proteins , Postural Balance/drug effects , Rats , Rats, Wistar , Reaction Time/drug effects , Reboxetine , Reserpine/pharmacology , Serotonin Plasma Membrane Transport Proteins , Venlafaxine Hydrochloride , p-Chloroamphetamine/pharmacology
4.
J Nucl Med ; 42(6): 932-7, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11390559

ABSTRACT

UNLABELLED: Ca(2+) channels play a key role in the basic working of the heart. There is one particular type of Ca(2+) channel in cardiac cells (L-type) whose gating is affected in different ways by beta-adrenoceptors and 1,4-dihydropyridines. In this study, we used ex vivo studies and PET to evaluate and compare the myocardial kinetics of the enantiomers labeled with (11)C (the more active: S12968, absolute configuration S; the less active: S12967, absolute configuration R) of the L-type Ca(2+) channel antagonist S11568 (3-ethyl 5-methyl (+/-)-2-[(2-(2-aminoethoxy)ethoxy) methyl]-4-(2,3-dichlorophenyl)-6-methyl-1,4-dihydropyridine-3,5-dicarboxylate). METHODS: [(11)C]S12968 was injected into the tail vein of rats (0.22 kBq--5.92 MBq) to assess the relationship between injected dose and myocardial uptake. A series of 5 rats was pretreated with 4 micromol unlabeled S12968 5 min before injection of 2.2 kBq [(11)C]S12968. In another series of 5 rats, unlabeled S12698 (4 micromol) was injected 5 min after injection of 2.2 kBq [(11)C]S12968. The animals were killed 15 min later, and the myocardial radioactivity was assessed in a gamma well counter. Beagle dogs received injections of 5-15 nmol [(11)C]S12968 or [(11)C]S12967 and were imaged with PET. Presaturation and displacement experiments using 2 micromol/kg unlabeled S12968 or 6 mol/kg S12967 were performed. RESULTS: In rats, a statistically significant relationship between myocardial uptake and injected dose of S12968 was observed. Pretreatment or displacement with unlabeled S12968 reduced myocardial radioactivity by 75% and 70%, respectively. In dogs, after injection of 5 nmol of each enantiomer, myocardial radioactivity plateaued within 3 min and the clearance from blood was rapid. Injection of 13--15 nmol [(11)C]S12968 led to a higher myocardial uptake and a more rapid washout, which were related to an increased coronary blood flow as shown by the linear relationship between k(1)--an estimate of coronary blood flow--and the mass of S12968 injected. Presaturation and displacement experiments showed that 70%--80% of S12968 binding was specific. This specificity was not observed with S12967. Plasma metabolite analysis showed that 70% of the compound was unchanged 20 min after injection. CONCLUSION: These results show the feasibility of imaging myocardial L-type Ca(2+) channels in vivo using [(11)C]S12968.


Subject(s)
Calcium Channel Blockers , Carbon Radioisotopes , Dihydropyridines , Heart/diagnostic imaging , Tomography, Emission-Computed , Animals , Calcium Channel Blockers/pharmacokinetics , Carbon Radioisotopes/pharmacokinetics , Coronary Circulation , Dihydropyridines/pharmacokinetics , Dogs , Female , Male , Myocardium/metabolism , Rats , Rats, Sprague-Dawley , Tissue Distribution
5.
J Pharmacol Exp Ther ; 292(1): 38-53, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10604930

ABSTRACT

S18327 displayed modest affinity for human (h)D(2) and hD(3) receptors and high affinity for hD(4) receptors. At each, S18327 antagonized stimulation of [(35)S]guanosine-5'-O-(3-thio)triphosphate binding by dopamine (DA). It also blocked activation of mitogen-activated protein kinase at hD(3) receptors. The affinity of S18327 at hD(1) and hD(5) sites was modest. S18327 showed pronounced affinity for human serotonin (h5-HT)(2A) receptors and human alpha(1A)-adrenergic receptors (hARs), at which it antagonized increases in intracellular Ca(2+) concentration levels elicited by 5-HT and norepinephrine (NE), respectively. S18327 presented significant affinity for halpha(2A)-ARs and antagonized NE-induced[(35)S]guanosine-5'-O-(3-thio)triphosphate binding both at these sites and at alpha(2)-ARs in rat amygdala. Reflecting blockade of alpha(2)-autoreceptors, S18327 enhanced firing of adrenergic neurons in locus ceruleus, accelerated hippocampal synthesis of NE, and increased dialysate levels of NE in hippocampus, accumbens, and frontal cortex. S18327 abolished inhibition of ventrotegmental area-localized dopaminergic neurons by apomorphine. However, S18327 alone did not affect their activity and only modestly enhanced cerebral turnover of DA and dialysate levels of DA in striatum and accumbens. In contrast, S18327 markedly increased dialysate levels of DA in frontal cortex, an action abolished by the selective alpha(2)-AR agonist, S18616. Finally, S18327 reduced synthesis and dialysate levels of 5-HT in striatum and suppressed firing of dorsal raphe-localized serotonergic neurons, an action attenuated by the alpha(1)-AR agonist cirazoline. In conclusion, S18327 possesses marked antagonist activity at alpha(1)-ARs and D(4) and 5-HT(2A) receptors and less potent antagonist activity at alpha(2)-ARs and D(1) and D(2) receptors. Antagonism by S18327 of alpha(2)-ARs enhances adrenergic transmission and reinforces frontocortical dopaminergic transmission, whereas blockade of alpha(1)-ARs inhibits dorsal raphe-derived serotonergic pathways. As further described in the accompanying paper, this profile of activity may contribute to the potential antipsychotic properties of S18327.


Subject(s)
Adrenergic Antagonists/pharmacology , Antipsychotic Agents/pharmacology , Brain/drug effects , Dopamine Antagonists/pharmacology , Imidazoles/pharmacology , Isoxazoles/pharmacology , Serotonin Antagonists/pharmacology , Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-2 Receptor Antagonists , Animals , Brain/metabolism , Brain/pathology , Calcium/metabolism , Dose-Response Relationship, Drug , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , In Vitro Techniques , Male , Norepinephrine/metabolism , Protein Binding , Rats , Rats, Wistar , Time Factors
6.
J Pharmacol Exp Ther ; 292(1): 54-66, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10604931

ABSTRACT

S18327 was dose-dependently active in several models of potential antipsychotic activity involving dopaminergic hyperactivity: inhibition of apomorphine-induced climbing in mice, of cocaine- and amphetamine-induced hyperlocomotion in rats, and of conditioned avoidance responses in rats. Furthermore, reflecting its high affinity at serotonin(2A) sites, S18327 potently blocked phencyclidine-induced locomotion and 1-[2, 5-dimethoxy-4-iodophenyl]-2-aminopropane-induced head-twitches in rats. In models of glutamatergic hypoactivity, S18327 blocked hyperlocomotion and spontaneous tail-flicks elicited by the N-methyl-D-aspartate antagonist dizocilpine. The actions of S18327, together with its binding profile at multiple monoaminergic receptors (15 parameters in total), were compared with those of clozapine, haloperidol, and 11 other antipsychotics by multiparametric analysis, and the resulting dendrogram positioned S18327 close to clozapine. Consistent with a clopazine-like profile, S18327 generalized to a clozapine discriminative stimulus and evoked latent inhibition in rats, blocked aggression in isolated mice, and displayed anxiolytic properties in the ultrasonic vocalization and Vogel procedures in rats. Relative to the above paradigms, only markedly (>20-fold) higher doses of S18327 were active in models predictive of potential extrapyramidal side effects: induction of catalepsy and prolactin secretion, and inhibition of methylphenidate-induced gnawing in rats. S18327 showed only modest affinity for histaminic and muscarinic receptors. Multiparametric analysis of these data distinguished S18327 from both haloperidol (high extrapyramidal potential) and clozapine (high histaminic and muscarinic affinity). In conclusion, S18327 displays a broad-based pattern of potential antipsychotic activity at doses appreciably lower than those eliciting extrapyramidal side effects. In this respect, S18327 closely resembles clozapine, but it is chemically distinct and displays weak affinity for histaminic and muscarinic receptors.


Subject(s)
Adrenergic Antagonists/pharmacology , Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Haloperidol/pharmacology , Imidazoles/pharmacology , Isoxazoles/pharmacology , Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-2 Receptor Antagonists , Aggression/drug effects , Amphetamine/pharmacology , Animals , Apomorphine/pharmacology , Avoidance Learning/drug effects , Cocaine/pharmacology , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Extrapyramidal Tracts/drug effects , Male , Mice , Pain Measurement , Protein Binding , Rats , Rats, Wistar , Receptors, Histamine/drug effects , Receptors, Muscarinic/drug effects
7.
J Pharmacol Exp Ther ; 287(1): 167-86, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9765336

ABSTRACT

The novel benzoindane S 18126 possessed > 100-fold higher affinity at cloned, human (h) D4 (Ki = 2.4 nM) vs. hD2 (738 nM), hD3 (2840 nM), hD1 (> 3000 nM) and hD5 (> 3000 nM) receptors and about 50 other sites, except sigma1 receptors (1.6 nM). L 745,870 similarly showed selectivity for hD4 (2.5 nM) vs. hD2 (905 nM) and hD3 (> 3000 nM) receptors. In contrast, raclopride displayed low affinity at hD4 (> 3000 nM) vs. hD2 (1.1 nM) and hD3 receptors (1.4 nM). Stimulation of [35S]-GTPgammaS binding at hD4 receptors by dopamine (DA) was blocked by S 18126 and L 745,870 with Kb values of 2.2 and 1.0 nM, respectively, whereas raclopride (> 1000 nM) was inactive. In contrast, raclopride inhibited stimulation of [35S]-GTPgammaS binding at hD2 sites by DA with a Kb of 1.4 nM, whereas S 18126 (> 1000 nM) and L 745,870 (> 1000 nM) were inactive. As concerns presynaptic dopaminergic receptors, raclopride (0.01-0.05 mg/kg s.c. ) markedly enhanced DA synthesis in mesocortical, mesolimbic and nigrostriatal dopaminergic pathways. In contrast, even high doses (2. 5-40.0 mg/kg s.c.) of S 18126 and L 745,870 were only weakly active. Similarly, raclopride (0.016 mg/kg i.v.) abolished inhibition of the firing rate of ventrotegmental dopaminergic neurons by apomorphine, whereas even high doses (0.5 mg/kg i.v.) of S 18126 and L 745,870 were only weakly active. As regards postsynaptic dopaminergic receptors, raclopride potently (0.01-0.3 mg/kg s.c.) reduced rotation elicited by quinpirole in rats with unilateral lesions of the substantia nigra, antagonized induction of hypothermia by PD 128, 907, blocked amphetamine-induced hyperlocomotion and was effective in six further models of potential antipsychotic activity. In contrast, S 18126 and L 745,870 were only weakly active in these models (5.0-> 40.0 mg/kg s.c.). In six models of extrapyramidal and motor symptoms, such as induction of catalepsy, raclopride was likewise potently active (0.01-2.0 mg/kg s.c.) whereas S 18126 and L 745,870 were only weakly active (10.0-80.0 mg/kg s.c.). In freely moving rats, raclopride (0.16 mg/kg s.c.) increased levels of DA by + 55% in dialysates of the frontal cortex. However, it also increased levels of DA in the accumbens and striatum by 70% and 75%, respectively. In contrast to raclopride, at a dose of 0.16 mg/kg s.c. , neither S 18126 nor L 745,870 modified frontal cortex levels of DA. However, at a high dose (40.0 mg/kg s.c.), S 18126 increased dialysate levels of DA (+ 85%) and noradrenaline (+ 100%), but not serotonin (+ 10%), in frontal cortex without affecting DA levels in accumbens (+ 10%) and striatum (+ 10%). In conclusion, S 18126 and L 745,870 behave as potent and selective antagonists of cloned, hD4 vs. other dopaminergic receptor types in vitro. However, their in vivo effects at high doses probably reflect residual antagonist actions at D2 (or D3) receptors. Selective blockade of D4 receptors was thus associated neither with a modification of dopaminergic transmission nor with antipsychotic (antiproductive) or extrapyramidal properties. The functional effects of selective D4 receptor blockade remain to be established.


Subject(s)
Dioxins/pharmacology , Dopamine Antagonists/pharmacology , Dopamine D2 Receptor Antagonists , Piperazines/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Salicylamides/pharmacology , Animals , Basal Ganglia Diseases/chemically induced , Body Temperature/drug effects , Brain/drug effects , Dopamine/analysis , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Male , Mice , Raclopride , Rats , Rats, Wistar , Receptors, Dopamine D4 , Receptors, sigma/drug effects , Serotonin/analysis
8.
Nucl Med Biol ; 25(4): 339-42, 1998 May.
Article in English | MEDLINE | ID: mdl-9639294

ABSTRACT

[11C]S11568 (3-ethyl-5-methyl 2-[2-(2-aminoethoxy)ethoxymethyl]-4-(2,3-dichlorophenyl)-6-methyl- 1,4-dihydropyridine-3,5-dicarboxylate) is a powerful ligand for the visualization of the cardiac calcium channel in vivo using PET. The aim of the present study was to synthesize a lipophilic, nonionized derivative of S11568 to facilitate its penetration into the brain. To increase the lipophilicity and to remove simultaneously the ionic nature of our ligand, the N-tert-butoxycarbonyl (N-Boc) derivative of S11568 was synthesized. An IC50 value of 1.7 nM for this derivative confirmed that both the affinity and selectivity for the calcium channel was unaltered by this chemical modification (S11568 with IC50 value of 9.9 nM). The biologically more active enantiomer of S11568, the levogyre isomer S12968, was labelled with 11C using [11C]iodomethane. The lipophilicity of the N-Boc derivative was increased by a factor of three to four when compared to the parent compound (as determined by the measurement of the octanol/buffer partition coefficients). In vivo, this derivative slightly crosses the blood-brain barrier, as demonstrated by a 4-fold increase (with respect to the parent compound S12968) of the radioactivity in the brain using the 11C-labelled N-Boc S12968. This uptake remained too low to be suitable for imaging calcium channels.


Subject(s)
Brain/metabolism , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/pharmacokinetics , Calcium Channels/metabolism , Dihydropyridines/chemical synthesis , Dihydropyridines/pharmacokinetics , Animals , Blood-Brain Barrier , Calcium Channel Blockers/chemistry , Dihydropyridines/chemistry , Male , Rats , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Tissue Distribution , Tomography, Emission-Computed
9.
J Pharmacol Exp Ther ; 282(1): 148-61, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9223550

ABSTRACT

In these studies, we characterized the influence of the novel benzodioxopiperazine serotonin (5-HT)1A ligand, S 15535, on the release of 5-HT in rat hippocampus and compared its potential anxiolytic properties with those of the 5-HT1A receptor partial agonist, buspirone, the 5-HT1A antagonist, WAY 100,635 and the benzodiazepine, diazepam (DZM). (Doses are in milligrams per kilogram s.c., unless otherwise specified.) S 15535 dose-dependently (0.3-3.0) reduced dialysate concentrations of 5-HT in the hippocampus of anesthetized rats. This action of S 15535 (3.0) was blocked by WAY 100,635 (0.3), (-)-penbutolol (2.0) and (-)-tertatolol (8.0), antagonists at 5-HT1A autoreceptors. In rats, fear-induced ultrasonic vocalizations (USVs) were dose-dependently abolished by S 15535 (0.16-2.5 s.c. and 0.63-10.0 p.o.), an action mimicked by buspirone (0.02-2.5) and DZM (0.16-10.0). Further, the action of S 15535 (0.63) was abolished by WAY 100,635 (0.16) and (-)-penbutolol (10.0), which were inactive alone. S 15535 dose-dependently (0.63-10.0 s.c. and 2.5-40.0 p.o.) blocked aggressive encounters in isolated mice; buspirone (0.16-10.0) and, at high doses, DZM (2.5-40.0) were also effective. WAY 100,635 (0.16), which was inactive alone, fully antagonized the antiaggressive actions of S 15535 (2.5). In an elevated plus-maze, neither S 15535 (0.0025-10.0), buspirone (0.0025-10.0) nor WAY 100,635 (0.00063-0.63) significantly increased open-arm entries, whereas they were increased by DZM (0.16-0.63). In the pigeon conflict test, S 15535 (0.04-0.16 i.m.) markedly increased punished responses and only slightly decreased unpunished responses, even at a 64-fold higher dose. In contrast, buspirone (0.16-2.5 i.m.) and DZM (0.04-2.5 i.m.) showed no or a less marked (4-fold) separation between doses increasing punished and decreasing unpunished responses. In the presence of the 5-HT1A antagonist, (-)-alprenolol (10.0 mg/kg i.m.), S 15535 did not increase punished responses. In a Geller conflict paradigm in rats, S 15535 dose dependently (0.3-3.0) increased punished responses, and its action (1.0) was blocked by (-)-penbutolol (8.0). S 15535 (0.63-40.0 s.c. and 2.5-40.0 p.o.) exerted little influence on motor behavior. In conclusion, in line with its net inhibition of serotoninergic transmission by activation of 5-HT1A autoreceptors and blockade of postsynaptic 5-HT1A receptors, S 15535 expresses anxiolytic activity. In addition, it displays antiaggressive (and antidepressant, accompanying paper) properties. Further, S 15535 does not compromise motor behavior at doses over which it expresses its anxiolytic properties. Thus, S 15535 represents a promising candidate for the treatment of anxious states in man.


Subject(s)
Anti-Anxiety Agents/pharmacology , Hippocampus/drug effects , Piperazines/pharmacology , Receptors, Serotonin/drug effects , Serotonin Receptor Agonists/pharmacology , Aggression/drug effects , Animals , Buspirone/pharmacology , Columbidae , Female , Hippocampus/metabolism , Male , Mice , Mice, Inbred ICR , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Serotonin, 5-HT1 , Vocalization, Animal/drug effects
10.
Bioorg Med Chem ; 5(4): 749-64, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9158874

ABSTRACT

(+/-)-S11568 (1, 3-ethyl-5-methyl-(+/-)-2-[(2-(2-aminoethoxy)ethoxy) methyl]-4-(2,3-dichlorophenyl)-6-methyl-1,4-dihydropyridine-3, 5-dicarboxylate), has an in vitro profile of high potency and of high selectivity for the low-voltage dependent. L-type calcium channel. In in vitro binding studies, it displaced specifically bound (-)-[3H]PN 200-110 (isradipine (2), the reference molecule for in vitro studies) from cardiac and vascular smooth muscle preparations with potencies of 5.6 and 51 nM, respectively. It also appears as a pure pharmacological antagonist acting at a single channel L-type and free of any interaction at the benzothiazepine binding site such as amlodipine (3). Both enantiomers of S11568 have in vitro activities, the dextro isomer S12967 ((+)-1) being 6 to 18-fold less potent than the levo one S12968 ((-)-1). Two couples of optically active labelling precursors of S11568, ((-)-10/(+)-10 and (-)-14/(+)-14) have been synthesized using a modified Hantzsch's dihydropyridine synthesis. In both cases, the enantiomers were separated by preparative chiral HPLC. They both have been independently labelled with carbon-11, using [11C]diazomethane or [11C]iodomethane to give multimilliCurie quantities of (-)-1 (S12968) and (+)-1 (S12967) with high specific activities (500-1000 mCi/mumol, 18.5-37.0 GBq/mumol). Both enantiomers appear suitable for PET experiments: their myocardial concentration increases after a bolus injection to reach a maximum in 2 min and then remains on a plateau with a slight downslope while the blood concentration falls rapidly. Myocardial uptake was threefold higher than lung uptake, leading to a good contrast on PET images. The present preliminary biological results obtained in Beagle dogs showed that both enantiomers have similar myocardial kinetics and in vivo affinity for the left ventricular myocardium.


Subject(s)
Calcium Channel Blockers/chemical synthesis , Dihydropyridines/chemical synthesis , Heart/diagnostic imaging , Tomography, Emission-Computed/methods , Amlodipine/chemistry , Amlodipine/metabolism , Animals , Binding, Competitive , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/metabolism , Calcium Channel Blockers/pharmacology , Carbon Radioisotopes , Chromatography, High Pressure Liquid , Dihydropyridines/chemistry , Dihydropyridines/metabolism , Dihydropyridines/pharmacology , Dogs , Heart/drug effects , Heart Diseases/diagnostic imaging , Isotope Labeling , Isradipine/chemistry , Isradipine/metabolism , Magnetic Resonance Spectroscopy , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocardium/metabolism , Reproducibility of Results , Stereoisomerism
11.
Eur J Pharmacol ; 325(1): 9-12, 1997 Apr 23.
Article in English | MEDLINE | ID: mdl-9151932

ABSTRACT

The mixed 5-HT2A/5-HT2B/5-HT2C receptor agonist, m-(chlorophenyl)piperazine (mCPP), elicited penile erections in rats, an action mimicked by the selective 5-HT2C receptor agonist, RO 60-0175 (S)-2-(6-chloro-5-fluoroindol-1-yl)-1-methylethylamine), whereas the preferential 5-HT2B receptor agonist, BW 723C86 (1-[5-(thienylmethoxy)-1H-3-indoyl] propan-2-amine) was ineffective. The actions of mCPP and RO 60-0175 were dose-dependently abolished by the novel 5-HT2B/5-HT2C receptor antagonists, SB 200,646 (1-(1-methylindol-5-yl)-3-(3-pyridyl) urea) and SB 206,553 (5 methyl-1-(3-pyridil-carbamoyl)-1,2,3,5-tetrahydropyrrolo[2,3 -f]indole). In contrast, penile erections were not significantly affected by the selective 5-HT2B receptor antagonist, SB 204,741 (1-(1-methylindol-5-yl)-3-(3-methylisothiazol-5-yl)-urea) nor by the selective 5-HT2A receptor antagonist, MDL 100,907 ([R(+)-alpha-(2,3-dimethoxyphenyl)-1-[2-(4-fluorophenylethyl)]-4-p iperidine-methanol]). These data provide rigorous pharmacological evidence that activation of 5-HT2C receptor elicits penile erections in the rat. This model should, thus, be of use for characterising novel ligands at this site.


Subject(s)
Penile Erection/drug effects , Penile Erection/physiology , Receptors, Serotonin/drug effects , Receptors, Serotonin/physiology , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Animals , Dose-Response Relationship, Drug , Ethylamines/pharmacology , Indoles/pharmacology , Male , Piperazines/pharmacology , Pyridines/pharmacology , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT2C , Thiophenes/pharmacology , Urea/analogs & derivatives , Urea/pharmacology
12.
Eur J Pharmacol ; 339(1): 43-51, 1997 Nov 19.
Article in English | MEDLINE | ID: mdl-9450615

ABSTRACT

The effects of the two isomers, (+)-S 16257 and (-)-S 16260, of a new bradycardic agent, (+/-)-S 15544 (7,8-dimethoxy 3-[3-[[(4.5-dimethoxybenzocyclobutan-1-yl)methyl] methylamino]propyl]1,3,4,5-tetrahydro-2H-3-benzazepin-2-one), were compared in vitro and in vivo on cardiac spontaneous rate and repolarization time. In the isolated rabbit sino-atrial node, the three compounds (3 microM) were equi-effective to reduce the action potential firing rate. In anesthetized pigs, both isomers (0.03, 0.1, 0.3 and 1 mg kg(-1) i.v.) were equipotent to reduce heart rate. For all compounds, the negative chronotropic effect resulted from a reduction in the slope of diastolic depolarization of pacemaker cells. In sino-atrial node cells, (-)-S 16260 (3 microM) increased action potential duration while (+)-S 16257 had a smaller effect. In driven guinea-pig papillary muscles exposed to increasing concentrations of compounds (0.1 to 10 microM) a small prolongation of action potential duration was observed. This prolongation was more marked in rabbit Purkinje fibers stimulated at a low rate. In all cardiac preparations the highest prolongation was observed with (-)-S 16260. In vivo, (-)-S 16260 prolonged QTc at the two highest doses tested while (+)-S 16257 had no effect. In conclusion, resolution of (+/-)-S 15544 into its two enantiomers yielded compounds with the same bradycardic effects. Of the isomers, (+)-S 16257 has an increased specificity with minimal direct effect on action potential repolarization.


Subject(s)
Benzazepines/pharmacology , Cardiotonic Agents/pharmacology , Sinoatrial Node/drug effects , Action Potentials/drug effects , Animals , Biological Clocks , Electrocardiography/drug effects , Guinea Pigs , Heart Rate/drug effects , Ivabradine , Male , Papillary Muscles/drug effects , Purkinje Fibers/drug effects , Rabbits , Stereoisomerism , Swine
13.
Eur Neuropsychopharmacol ; 5(4): 481-9, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8998401

ABSTRACT

The binding and locomotor profile of a new sigma ligand, S14905, (isobutyl-N-(1-indan-2yl-piperid-4-yl)N-methyl carbamate, furamate) was studied. The binding data revealed that S14905 has a high affinity for sigma receptors and very low affinity for both dopamine D1 and D2 receptors. We have demonstrated that this sigma ligand prevents the locomotor stimulation induced by morphine (32 and 64 mg/kg), cocaine (16 mg/kg), amphetamine (4 mg/kg) and adrafinil (32 mg/kg) at doses lower than those required to depress spontaneous locomotor activity. The antagonism observed in the present study seems to be more specific of morphine induced hyperlocomotion. The high affinity of this compound for sigma receptors makes it a good choice to study the role of this receptor in the CNS. In addition, S14905 does not directly block dopamine receptors but may modulate them in some manner, and would thus warrant further study as a potential atypical antipsychotic agent, and an antagonist for the hyperactivity induced by opiate drug.


Subject(s)
Central Nervous System Stimulants/pharmacology , Indans/pharmacology , Motor Activity/drug effects , Piperidines/pharmacology , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Cocaine/antagonists & inhibitors , Cocaine/pharmacology , Dose-Response Relationship, Drug , Hydroxamic Acids/antagonists & inhibitors , Hydroxamic Acids/pharmacology , Male , Mice , Morphine/pharmacology , Narcotics/pharmacology , Radioligand Assay , Receptors, sigma/drug effects , Stimulation, Chemical
14.
Neuropharmacology ; 34(12): 1693-6, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8788967

ABSTRACT

The selective dopamine D3 receptor antagonist [3H](+)S 14297 ((+)-[7-(N,N-dipropylamino)-5,6,7,8-tetrahydro-naphtho(2,3b)dihydro,2,3- furane]), labelled to high specific activity (145 Ci/mmol), bound to cloned human dopamine D3 receptors but displayed negligible binding to cloned human D2 receptors. [3H](+)S 14297 exhibited rapid association and dissociation, high affinity saturable binding (Kd = 7.0 nM) and a competition binding profile highly correlated with that of [125I]iodosulpride (r = 0.98).


Subject(s)
2-Naphthylamine/analogs & derivatives , Furans/metabolism , Receptors, Dopamine D2/metabolism , 2-Naphthylamine/metabolism , Animals , Binding, Competitive , CHO Cells , Cricetinae , Dopamine Agents/metabolism , Humans , Ligands , Receptors, Dopamine D3 , Sulpiride/analogs & derivatives , Sulpiride/metabolism , Transfection , Tritium
15.
J Pharmacol Exp Ther ; 275(2): 885-98, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7473180

ABSTRACT

Using [125I]-iodosulpride as a radioligand, the novel naphthofurane, (+/-)-S 11566 [(+/-)-[7-(N,N-dipropylamino)-5,6,7,8-tetra-hydro- naphtho(2,3b)dihydro,2,3-furane]) showed a marked preference for human, recombinant D3 as compared with D2 receptors stably transfected into Chinese hamster ovary cells (Kis = 24/529 nM). This activity resided in its (+)-eutomer, (+)-S 14297 (13/297 nM) as compared with its (-)-distomer, (-)-S 17777 (406/3544 nM). In contrast, (+)-AJ 76 manifested only a mild 2-fold preference for D3 sites (70/154 nM), whereas haloperidol and six additional antagonists showed a mild (2-7-fold) preference for D2 sites. As concerns agonists, (+)-7-OH-DPAT, (+/-)-CGS 15855A, quinelorane, (-)-quinpirole and N-0434 displayed a preference (6-40-fold) for D3 receptors, whereas piribedil showed a slight, 2-fold, preference for D2 sites (243/126 nM). (+)-S 14297 showed low (> 1.0 microM) affinity at rat D1 and D2 sites and at cloned, human D4 and D5 receptors and only low affinity (145 to > 10,000 nM) at all other sites examined. In vivo, administered s.c., (+)-7-OH-DPAT, CGS 15855A, quinelorane, (-)-quinpirole and N-0434 potently evoked hypothermia. Across all (8) agonists tested, potency correlated significantly with affinity at D3 sites (r = .84, P < .001) but not D2 sites (r = .50, P > .05). (+)-S 14297 (0.16-1.25 mg/kg, s.c.) blocked the induction of hypothermia by (+)-7-OH-DPAT, CGS 15855A and (-)-quinpirole, but not by the alpha 2-adrenergic agonist, clonidine, without influencing core temperature alone. In contrast, (-)-S 17777 (10.0 mg/kg, s.c.) was only partially active. Across all (9) antagonists, potency for inhibition of (+)-7-OH-DPAT-induced hypothermia correlated more strongly with affinity at D3 (r = .96, P < .001) than D2 (r = .75, P < .02) sites. Whereas haloperidol and the other antagonists provoked prolactin secretion and elicited catalepsy, (+)-S 14297 and (+/-)-S 11566 at doses of up to 10.0 and 40.0 mg/kg, s.c., respectively, were not significantly effective (P > .05). Across all antagonists, potency for eliciting prolactin secretion and catalepsy correlated better with affinity at D2 (r = .95 and .96) than D3 (r = .76 and .91) sites. In conclusion, these data demonstrate that the novel naphtofurane, (+)-S 14297, is a selective ligand (antagonist) at dopamine D3 receptors and suggest that their activation mediates hypothermia in the rat.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
2-Naphthylamine/analogs & derivatives , Catalepsy/etiology , Dopamine Antagonists/pharmacology , Furans/pharmacology , Prolactin/metabolism , Receptors, Dopamine D2/physiology , Receptors, Dopamine/physiology , 2-Naphthylamine/pharmacology , Animals , Body Temperature Regulation/drug effects , CHO Cells , Cricetinae , Humans , Male , Rats , Rats, Wistar , Receptors, Dopamine/classification , Receptors, Dopamine/drug effects , Recombinant Proteins , Stereoisomerism
16.
J Pharmacol Exp Ther ; 275(2): 899-913, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7473181

ABSTRACT

The preferential dopamine (DA) D3 versus D2 receptor agonist, (+)-7-OH-DPAT, dose-dependently decreased DA synthesis in the nucleus accumbens, olfactory tubercles, striatum and frontal cortex. This action was potently mimicked by several other high-potency D3 agonists: CGS 15855A, (-)-quinpirole, quinelorane and N-0434. In contrast, piribedil, which displays a mild preference for D2 sites, was less active. Across eight agonists, potency for inhibition of DA synthesis correlated more potently to affinity at D3 (r = .82 +/- .04) than D2 receptors (r = .60 +/- .06, P < .05). Correlations were also marked to potency for induction of a further D3-mediated response, hypothermia (r = .93 +/- .02). The novel and selective D3 versus D2 antagonist, (+/-)-S 11556, attenuated the action of (+)-7-OH-DPAT in each structure. This action was shared by its active (+)-eutomer, (+)-S 14297, whereas its inactive (-)-distomer, (-)-S 17777, was ineffective. (+)-S 14297 similarly attenuated the inhibitory action of CGS 15855A and (-)-quinpirole upon DA synthesis, whereas it failed to modify inhibition of striatal DA synthesis by the alpha 2-adrenergic receptor agonist, clonidine. As compared with the D2/D3 receptor antagonist, haloperidol, neither (+/-)-S 11566 nor (+)-S 14297 modified DA turnover upon administration alone. Furthermore, across (nine) antagonists, potency in facilitating DA synthesis more powerfully correlated to affinity at D2 (r = .94 +/- .01) than D3 (r = .73 +/- .01) sites (P < .01). Correlations were also marked to potency for induction of catalepsy (r = .91 +/- .01) and prolactin secretion (r = .89 +/- .01) but not for antagonism of (+)-7-OH-DPAT-induced hypothermia (r = .60 +/- .01). In freely moving rats, (+)-7-OH-DPAT dose-dependently reduced dialysate concentrations of DA in the nucleus accumbens and contralateral striatum: this action was potently mimicked by CGS 15855A, but only weakly so by piribedil. (+)-S 14297 markedly attenuated the action of (+)-7-OH-DPAT, whereas (-)-S 17777 was inactive. In contrast, haloperidol completely blocked the action of (+)-7-OH-DPAT. Finally, in distinction to haloperidol, upon administration alone, (+)-S 14297 did not significantly enhance the release of DA. In conclusion, these data suggest that D3 (auto)receptors control synthesis and release of DA in dopaminergic pathways innervating the limbic system, cortex and striatum.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
2-Naphthylamine/analogs & derivatives , Brain/physiology , Dopamine Antagonists/pharmacology , Dopamine/metabolism , Furans/pharmacology , Receptors, Dopamine/physiology , 2-Naphthylamine/pharmacology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Brain/drug effects , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Dopamine Agonists/pharmacology , Male , Nucleus Accumbens/metabolism , Olfactory Bulb/metabolism , Rats , Rats, Wistar , Receptors, Dopamine/classification , Receptors, Dopamine D2/physiology , Tetrahydronaphthalenes/pharmacology
17.
J Med Chem ; 38(20): 4044-55, 1995 Sep 29.
Article in English | MEDLINE | ID: mdl-7562940

ABSTRACT

Benzocycloalkyl and benzocycloalkenyl moities linked, directly or via an alkyl chain, to oxygen-bearing heteroarylpiperazines were synthesized, in an attempt to obtain potent and selective antagonists at postsynaptic 5-HT1A receptors. From the numerous arylpiperazines described in the literature, 1-(2,3-dihydro-1,4-benzodioxin-5-yl)piperazine (3a) was chosen as a model of an arylpiperazine in view of its selectivity for 5-HT1A receptors versus alpha 1-, alpha 2-, and beta-adrenergic receptors, as well as dopamine D1 and D2 receptors. Two other closely-related arylpiperazines, 1-(1,5-benzodioxepin-6-yl)piperazine (3b) and 1-(benzofuran-7-yl)piperazine (3c), were also examined in this study. All compounds showed high affinity at 5-HT1A sites (8.10 < or = pKis < or = 9.35), and the majority behaved as antagonists in vivo in blocking the hypothermia induced by the 5-HT1A agonist 8-OH-DPAT in the absence of a marked effect alone at equivalent doses. An in vivo evaluation of dopamine D2 receptor antagonist properties revealed that the majority of compounds was devoid of activity at this site, in marked contrast to BMY 7378 which displayed virtually no selectivity for 5-HT1A versus dopamine D2 receptors. Moreover, six compounds of the present series, 8, 10, 11, 14, 25, and 37, showed > 10-fold selectivity in vitro for 5-HT1A versus alpha 1-adrenergic receptors. Compound 14 displayed an optimal compromise between potency (pKi = 8.75), marked antagonist activity, and selectivity toward alpha 1-adrenergic (81-fold) and dopamine D2 (195-fold) receptors. These characteristics clearly distinguish 14 from previously-reported ligands such as the postsynaptic 5-HT1A antagonist BMY 7378 and the weak partial agonist NAN 190 which, in contrast to the compounds of this series, belong to the well-exemplified class of imido derivatives of (o-methoxyphenyl)piperazines. The availability of 14 (S 15535) should facilitate the further elucidation of the functional role and potential therapeutic significance of 5-HT1A receptors.


Subject(s)
Piperazines/pharmacology , Serotonin Antagonists/pharmacology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Male , Rats , Rats, Wistar , Structure-Activity Relationship
18.
Eur J Pharmacol ; 265(3): 175-7, 1994 Nov 24.
Article in English | MEDLINE | ID: mdl-7875234

ABSTRACT

In Chinese hamster ovary cells stably transfected with recombinant rat dopamine D2 or D3 receptors, the naphthofurane antagonist, (+)-S 14297 [(+)-[7-(N,N-dipropylamino)-5,6,7,8-tetrahydro- naphtho(2,3b)dihydro,2,3-furane]], displayed a pronounced preference for dopamine D3 versus D2 receptors: Ki values = 13 and 365 nM, respectively. In contrast, its distomer, (-)-S 17777, showed low affinity (296 versus 3403 nM). The aminotetralin agonist, (+)-7-OH-DPAT (7-hydroxy-2-(di-n-propylamino)tetralin), also showed high affinity at dopamine D3 (1.8 nM) versus D2 (96 nM) receptors while its (-)-isomer showed low affinity (71 and 1461 nM). In freely moving rats, (+)-7-OH-DPAT (0.16 mg/kg s.c.)-but not (-)-7-OH-DPAT-decreased dialysate levels of dopamine in the nucleus accumbens. (+)-S 14297 (1.25 mg/kg s.c.) markedly inhibited the action of (+)-7-OH-DPAT without influencing dopamine levels alone. Further, this action was stereospecific in that (-)-S 17777 (20.0 mg/kg s.c.) was inactive. In conclusion, data obtained with the novel, selective dopamine D3 receptor antagonist, (+)-S 14297 suggest that dopamine D3 autoreceptors modulate the release of dopamine from mesolimbic dopaminergic neurones.


Subject(s)
2-Naphthylamine/analogs & derivatives , Autoreceptors/physiology , Brain/drug effects , Dopamine Antagonists/pharmacology , Dopamine/metabolism , Furans/pharmacology , Receptors, Dopamine D2 , Receptors, Dopamine/physiology , 2-Naphthylamine/pharmacology , Animals , Autoreceptors/antagonists & inhibitors , Brain/metabolism , CHO Cells , Cricetinae , Cricetulus , Dopamine Agonists/pharmacology , Drug Interactions , Female , Male , Rats , Rats, Wistar , Receptors, Dopamine/drug effects , Receptors, Dopamine D3 , Tetrahydronaphthalenes/pharmacology
19.
Eur J Pharmacol ; 260(2-3): R3-5, 1994 Aug 01.
Article in English | MEDLINE | ID: mdl-7988633

ABSTRACT

The selective dopamine D3 receptor agonist, 7-OH-DPAT ((+)-7-hydroxy-2-(di-n-propylamino)tetralin) and the novel naphthofurane, S 14297 ((+)-[7-(N,N-dipropylamino)-5,6,7,8-tetrahydro- naphtho(2,3b)dihydro,2,3-furane]), bound with high affinity and selectivity to recombinant, human dopamine D3 versus D2 receptors stably transfected into Chinese hamster ovary cells: Ki values = 2 versus 103 nM for 7-OH-DPAT and 13 versus 297 nM for S 14297. In contrast, the putative dopamine D3 receptor antagonist, AJ 76 (cis-(+)-5-methoxy-1-methyl-2-(n- propylamino)tetralin), displayed low affinity and selectivity for dopamine D3 versus D2 sites (70 versus 154 nM). 7-OH-DPAT (0.01-0.16 mg/kg s.c.) provoked hypothermia in rats, an action abolished by S 14297 (0.04-0.63 mg/kg s.c.) and, less potently, by AJ 76 (0.16-2.5 mg/kg s.c.). S 14297 (20.0 mg/kg s.c.) did not modify prolactin secretion. These data suggest that dopamine D3 receptors mediate hypothermia in the rat and that S 14297 acts as a selective antagonist at these sites.


Subject(s)
2-Naphthylamine/analogs & derivatives , Body Temperature/drug effects , Furans/pharmacology , Hypothermia/drug therapy , Receptors, Dopamine D2 , Receptors, Dopamine/metabolism , Tetrahydronaphthalenes/toxicity , 2-Naphthylamine/administration & dosage , 2-Naphthylamine/metabolism , 2-Naphthylamine/pharmacology , 2-Naphthylamine/therapeutic use , Animals , Binding, Competitive , CHO Cells/drug effects , CHO Cells/metabolism , Cloning, Molecular , Cricetinae , Cricetulus , Furans/administration & dosage , Furans/metabolism , Furans/therapeutic use , Humans , Hypothermia/chemically induced , Ligands , Male , Rats , Rats, Wistar , Receptors, Dopamine/chemistry , Receptors, Dopamine D3 , Recombinant Proteins/metabolism , Tetrahydronaphthalenes/metabolism , Transfection
20.
Br J Pharmacol ; 112(1): 37-42, 1994 May.
Article in English | MEDLINE | ID: mdl-8032660

ABSTRACT

1. S 16257 is a new bradycardic agent. Its electropharmacological profile has been compared to that of the known bradycardic compound UL-FS 49 (Zatebradine). Intracellular recordings of action potentials (APs) were performed with conventional glass microelectrodes. 2. In the rabbit isolated sino-atrial node (SAN) tissue, S 16257 and UL-FS 49 (1 microM, 3 microM and 10 microM) were equipotent in slowing spontaneous APs firing predominantly by decreasing the rate of diastolic depolarization (at 3 microM, -23.8 +/- 3.9% and -27.9 +/- 2.6%, respectively). For the two compounds a maximal effect was obtained at 3 microM. In these preparations, action potential duration at 50% of total repolarization (APD50) was more affected by UL-FS 49 than S 16257 at any concentration tested (at 3 microM, +8.9 +/- 2.9% and +29.1 +/- 3.7% for S 16257 and UL-FS 49, respectively; P < or = 0.01). 3. To estimate the direct effects on AP duration, driven cardiac preparations were exposed to these agents. In guinea-pig papillary muscles, paced at a frequency of 1 Hz, increasing concentrations of S 16257 or UL-FS 49 (0.1 to 10 microM, 30 min exposure for each concentration) slightly prolonged AP repolarization. This prolongation was more marked for UL-FS 49 (at 1 microM, +6.1 +/- 0.6% and +11.2 +/- 1.3% elevation of APD50, for S 16257 and UL-FS 49, respectively). 4. Application of UL-FS 49 (3 microM) to rabbit Purkinje fibres, triggered at a frequency of 0.25 Hz, induced a marked prolongation of APD50 and APD90 (+149.4 +/- 51.2% and +86.0 +/- 15.4%, respectively). S 16257 (3 MicroM) induced only a weak prolongation of AP (+ 14.1 +/- 5.0% and + 14.8 +/- 3.3% for APD50 and APD90, respectively) significantly smaller than in the case of UL-FS 49.5. These results show that S 16257 slows the rate of spontaneous AP firing in isolated SAN mainly by a reduction of the diastolic depolarization of the cells, which suggests an inhibition of the pace-maker current (If). S 16257 and UL-FS 49 are equipotent in their bradycardic effect but S 16257 is more specific as it induces less increase in myocardial repolarization time.


Subject(s)
Benzazepines/pharmacology , Cardiotonic Agents/pharmacology , Cardiovascular Agents/pharmacology , Heart/drug effects , Sinoatrial Node/drug effects , Action Potentials/drug effects , Animals , Cardiac Pacing, Artificial , Electrophysiology , Guinea Pigs , Heart/innervation , Heart Rate/drug effects , In Vitro Techniques , Ivabradine , Male , Papillary Muscles/drug effects , Purkinje Fibers/drug effects , Rabbits , Sinoatrial Node/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...