Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
J Mol Endocrinol ; 30(3): 317-29, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12790802

ABSTRACT

Peroxisome proliferator activated receptor (PPAR)-alpha controls the expression of multiple genes involved in lipid metabolism, and activators of PPAR-alpha, such as fibrates, are commonly used drugs in the treatment of hypertriglyceridemia and other dyslipidemic states. Recent data have also suggested a role for PPAR-alpha in insulin resistance and glucose homeostasis. In the present study, we have assessed the transcriptional and physiological responses to PPAR-alpha activation in a diet-induced rat model of insulin resistance. The two PPAR-alpha activators, fenofibrate and Wy-14643, were dosed at different concentrations in high-fat fed Sprague-Dawley rats, and the transcriptional responses were examined in liver using cDNA microarrays. In these analyses, 98 genes were identified as being regulated by both compounds. From this pool of genes, 27 correlated to the observed effect on plasma insulin, including PPAR-alpha itself and the leukocyte antigen-related protein tyrosine phosphatase (PTP-LAR). PTP-LAR was downregulated by both compounds, and showed upregulation as a result of the high-fat feeding. This regulation was also observed at the protein level. Furthermore, downregulation of PTP-LAR by fenofibric acid was demonstrated in rat FaO hepatoma cells in vitro, indicating that the observed regulation of PTP-LAR by fenofibrate and Wy-14643 in vivo is mediated as a direct effect of the PPAR agonists on the hepatocytes. PTP-LAR is one of the first genes involved in insulin receptor signaling to be shown to be regulated by PPAR-alpha agonists. These data suggest that factors apart from skeletal muscle lipid supply may influence PPAR-alpha-mediated amelioration of insulin resistance.


Subject(s)
Fenofibrate/pharmacology , Insulin Resistance , Liver/metabolism , Pyrimidines/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists , Transcription, Genetic , Animals , Blotting, Western , Liver/drug effects , Rats , Rats, Sprague-Dawley
2.
J Pharmacol Exp Ther ; 298(3): 1260-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11504829

ABSTRACT

Two dimeric analogs of the muscarinic acetylcholine receptor (mAChR) agonist phenylpropargyloxy-1,2,5-thiadiazole-quinuclidine (NNC 11-1314) were synthesized and pharmacologically evaluated. In radioligand binding assays on Chinese hamster ovary (CHO) cell membranes expressing the individual human M(1) to M(5) mAChR subtypes, both dimers [(3S)-1,4-bis-(3-[(3-azabicyclo[2.2.2]octanyl)-1,2,5-thiadiazol-4-yloxy]-1-propyn-1-yl)benzene,2-L-(+)-tartrate (NNC 11-1607) and (3S)-1,3-bis-(3-[(3-azabicyclo[2.2.2]octanyl)-1,2,5-thiadiazol-4-yloxy]-1-propyn-1-yl)benzene,2-L-(+)-tartrate (NNC 11-1585)] exhibited higher binding affinities than the monomeric NNC 11-1314. Only NNC 11-1585, however, displayed significant selectivity for the M(1) and M(2) mAChRs relative to the other subtypes. Although binding studies in rat brain homogenates supported the selectivity profile of NNC 11-1585 observed in the CHO membranes, rat heart membrane experiments revealed complex binding behavior for all three agonists that most likely reflected differences in species and host cell environment between the heart and CHO cells. Subsequent functional assays with phosphatidylinositol hydrolysis revealed that all three novel ligands were partial agonists relative to the full agonist oxotremorine-M at the CHO M(1), M(3), and M(5) mAChRs, with NNC 11-1607 displaying the highest functional selectivity. In the CHO M(2) and M(4) mAChR cells, agonist-mediated effects on forskolin-stimulated cAMP accumulation were characterized by bell-shaped concentration-response curves, with the exceptions of NNC 11-1607, which had no discernible effects at the M(2) mAChR, and NNC 11-1585, which could only inhibit cAMP accumulation at the M(4) mAChR. Thus, we identified NNC 11-1607 as a novel functionally selective M(1)/M(4) mAChR agonist. Our data suggest that dimerization of mAChR agonists is a viable approach in designing more potent and functionally selective agonists, as well as in providing novel tools with which to probe the nature of agonism at these receptors.


Subject(s)
Muscarinic Agonists/chemical synthesis , Muscarinic Agonists/pharmacology , Quinuclidines/pharmacology , Receptors, Muscarinic/drug effects , Thiadiazoles/pharmacology , Algorithms , Animals , Binding, Competitive/drug effects , CHO Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Cyclic AMP/metabolism , Ligands , Magnetic Resonance Spectroscopy , Phosphatidylinositols/metabolism , Structure-Activity Relationship
3.
Schizophr Res ; 49(1-2): 193-201, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11343877

ABSTRACT

(5R,6R)-6-(3-butylthio-1,2,5-thiadiazol-4-yl)-1-azabicyclo[3.2.1]octane) (BuTAC) is a novel, selective muscarinic receptor ligand with partial agonist mode of action at muscarinic M2 and M4 and antagonist mode of action at M1, M3 and M5 receptor subtypes in cloned cell lines. BuTAC exhibits functional dopamine receptor antagonism despite its lack of affinity for dopamine receptors, and parasympathomimetic effects in mice are produced only at doses well beyond the doses exhibiting the antipsychotic-like effects. In the present study we investigated the effects of BuTAC and the antipsychotic compounds clozapine, sertindole and olanzapine using one trial passive avoidance with mice as a model of learning and memory. Pharmacologically relevant doses of BuTAC and reference antipsychotics were identified, based on inhibition of apomorphine-induced climbing in mice as an assay measuring antidopaminergic potency. When ratios between the minimum effective dose (MED) for impairment of retention in passive avoidance and the MED for inhibition of apomorphine-induced climbing were calculated, BuTAC displayed a high ratio of >10, compared with clozapine (0.3), sertindole (3) and olanzapine (3). These data suggest that BuTAC is a potential novel antipsychotic which may have favourable effects on aspects of learning and memory.


Subject(s)
Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Avoidance Learning/drug effects , Disease Models, Animal , Memory/drug effects , Receptors, Muscarinic/drug effects , Schizophrenia/drug therapy , Animals , Behavior, Animal/drug effects , Male , Mice
4.
Eur J Pharmacol ; 402(3): 241-6, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10958890

ABSTRACT

(5R,6R)-6-(3-Propylthio-1,2,5-thiadiazol-4-yl)-1-azabicyclo[ 3.2.1]octane (PTAC) is a selective muscarinic receptor ligand. The compound exhibits high affinity for central muscarinic receptors with partial agonist mode of action at muscarinic M(2) and M(4) and antagonist mode of action at muscarinic M(1), M(3) and M(5) receptor subtypes. The compound was earlier reported to exhibit functional dopamine receptor antagonism in rodents despite its lack of affinity for dopamine receptors. In the present study, we report that PTAC, as well as the muscarinic receptor agonists pilocarpine and oxotremorine, dose-dependently decreased rates of intravenous self-administration (fixed ratio 1) of the indirect dopamine receptor agonist cocaine in drug naive mice. Similar decreases in cocaine self-administration rates were obtained with the dopamine receptor antagonists olanzapine, clozapine, risperidone, fluphenazine and haloperidol. These findings suggest that compounds with partial muscarinic receptor agonist mode of action may be used in the medical treatment of cocaine abuse.


Subject(s)
Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Muscarinic Agonists/pharmacology , Animals , Antipsychotic Agents/pharmacology , Bridged Bicyclo Compounds/pharmacology , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Male , Mice , Oxotremorine/pharmacology , Pilocarpine/pharmacology , Self Administration , Thiadiazoles/pharmacology
5.
Brain Res ; 870(1-2): 126-34, 2000 Jul 07.
Article in English | MEDLINE | ID: mdl-10869509

ABSTRACT

Previous studies indicate cholinergic systems suppress somatic nociception. The present studies determined if cholinergic muscarinic systems suppress visceral nociception, specifically, chemical irritation of the lower urinary tract. Bladders of urethane-anesthetized rats were cannulated through the dome for continuous-infusion cystometrogram recordings. EMG electrodes recorded anal sphincter activity. Infusion of 0.5% acetic acid into the bladder to produce irritation increased bladder activity and anal sphincter activity (i.e. activation of a nociceptive vesicoanal reflex). Oxotremorine (a muscarinic agonist) and (-)butylthio[2.2.2] (a mixed muscarinic agonist/antagonist) dose-dependently inhibited vesicoanal reflex activity. This inhibition was antagonized by atropine (a centrally active muscarinic antagonist) but not by scopolamine methylbromide (a peripherally restricted muscarinic antagonist). Physostigmine (a centrally active cholinesterase inhibitor) also dose-dependently inhibited vesicoanal reflex activity in an atropine-sensitive manner, while neostigmine (a peripherally restricted cholinesterase inhibitor) did not. Atropine alone (i.e. administered without prior administration of muscarinic agonist or cholinesterase inhibitor) produced robust but transient (15 min) increases in vesicoanal activity and bladder activity under conditions of acetic acid infusion into the bladder. Under conditions of saline infusion into the bladder, atropine had the opposite effect on bladder activity (i.e. inhibition). These studies indicate that an endogenous cholinergic muscarinic system can be activated by lower urinary tract irritation to suppress visceral nociception through central nervous system mechanisms.


Subject(s)
Cholinergic Agents/pharmacology , Muscarinic Antagonists/pharmacology , Nociceptors/physiology , Thiadiazoles/pharmacology , Urinary Bladder/physiology , Acetylcholine/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Atropine/pharmacology , Cholinesterase Inhibitors/pharmacology , Electromyography , Female , Muscarinic Agonists/pharmacology , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Neostigmine/pharmacology , Nociceptors/drug effects , Oxotremorine/pharmacology , Pain/physiopathology , Physostigmine/pharmacology , Rats , Rats, Sprague-Dawley , Reflex/drug effects , Reflex/physiology , Urinary Bladder/drug effects , Vasodilator Agents/pharmacology
6.
Schizophr Res ; 42(3): 249-59, 2000 May 05.
Article in English | MEDLINE | ID: mdl-10785583

ABSTRACT

Xanomeline is an M(1)/M(4) preferring muscarinic receptor agonist which decreased psychotic behaviors in patients with Alzheimer's disease, suggesting that xanomeline might be useful in the treatment of psychotic symptoms in patients with schizophrenia. The purpose of the present studies was, therefore, to compare the pharmacologic profile of xanomeline with that of known antipsychotic drugs. Electrophysiologically, xanomeline, after both acute and chronic administration in rats, inhibited A10 but not A9 dopamine cells in a manner which was blocked by the muscarinic receptor antagonist scopolamine. Behaviorally, xanomeline, like haloperidol, clozapine and olanzapine, blocked dopamine agonist-induced turning in unilateral 6-hydroxydopamine-lesioned rats, as well as apomorphine-induced climbing in mice. However, unlike the dopamine antagonist antipsychotic haloperidol, xanomeline did not produce catalepsy in rats. Moreover, xanomeline, like haloperidol, clozapine and olanzapine, inhibited conditioned avoidance responding in rats, an effect which also was blocked by scopolamine. The present results thus demonstrate that xanomeline has a pharmacologic profile which is similar to that of the atypical antipsychotics clozapine and olanzapine, thus indicating that xanomeline has the potential to be a novel approach in the treatment of psychotic symptoms in patients with schizophrenia.


Subject(s)
Antipsychotic Agents/pharmacology , Cholinergic Agonists/pharmacology , Muscarinic Agonists/pharmacology , Pyridines/pharmacology , Receptors, Cholinergic/drug effects , Thiadiazoles/pharmacology , Alzheimer Disease/psychology , Animals , Brain/physiology , Conditioning, Psychological/drug effects , Disease Models, Animal , Dopamine/metabolism , Electrophysiology , Male , Mice , Mice, Inbred Strains , Neurons/physiology , Psychotic Disorders/drug therapy , Psychotic Disorders/etiology , Rats , Rats, Sprague-Dawley , Schizophrenia/complications
7.
Am J Physiol ; 277(1): E93-102, 1999 07.
Article in English | MEDLINE | ID: mdl-10409132

ABSTRACT

Increased insulinotropic activity by the cholinergic agonist carbachol exists in insulin-resistant high fat-fed C57BL/6J mice. We examined the efficiency and potency of carbachol to potentiate glucose-stimulated insulin secretion and to improve glucose tolerance in these animals. Intravenous administration of carbachol (at 15 and 50 nmol/kg) markedly potentiated glucose (1 g/kg)-stimulated insulin secretion in mice fed both a control and a high-fat diet (for 12 wk), with a higher relative potentiation in high fat-fed mice measured as increased (1-5 min) acute insulin response and area under the 50-min insulin curve. Concomitantly, glucose tolerance was improved by carbachol. In fact, carbachol normalized glucose-stimulated insulin secretion and glucose tolerance in mice subjected to a high-fat diet. Carbachol (>100 nmol/l) also potentiated glucose-stimulated insulin secretion from isolated islets with higher efficiency in high fat-fed mice. In contrast, binding of the muscarinic receptor antagonist [N-methyl-(3)H]scopolamine to islet muscarinic receptors and the contractile action of carbachol on ileum muscle strips were not different between the two groups. We conclude that carbachol normalizes glucose tolerance in insulin resistance.


Subject(s)
Cholinergic Agonists/pharmacology , Dietary Fats/administration & dosage , Glucose Intolerance/metabolism , Glucose/pharmacology , Insulin/metabolism , Animals , Blood Glucose/analysis , Carbachol/metabolism , Carbachol/pharmacology , Dietary Fats/pharmacology , Female , Gastrointestinal Motility/physiology , Glucose Tolerance Test , Ileum/drug effects , In Vitro Techniques , Insulin/blood , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Reference Values
8.
J Pharmacol Exp Ther ; 290(2): 901-7, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10411607

ABSTRACT

The purpose of our studies was to determine the effects of muscarinic receptor agonists on conditioned avoidance responding in the rat. Rats were trained to avoid or escape an electric shock delivered to the feet in a discrete trial procedure. The muscarinic receptor agonists pilocarpine and [2-ethyl-8-methyl-2,8-diazaspiro(4. 5)decane-1,3-dione] hydrochloride (RS86) and the cholinesterase inhibitor physostigmine all decreased the percentage of avoidance responses at doses that produced less than approximately 30% response failures. Similar results were obtained with the antipsychotic drugs haloperidol, trifluoperazine, chlorpromazine, and clozapine. However, the benzodiazepine anxiolytic diazepam did not decrease avoidance responding up to doses that produced ataxia. On the other hand, oxotremorine and arecoline decreased avoidance responding only by producing response failures, whereas aceclidine produced intermediate changes. The muscarinic receptor antagonists scopolamine, trihexyphenidyl, and benztropine were without effect when administered alone but antagonized the decreases in avoidance responding produced by pilocarpine and RS86. Scopolamine had little effect on the decreases in avoidance responding produced by haloperidol. The newer muscarinic receptor partial agonists or agonist/antagonists [R-(Z)-(+)-alpha-(methoxyimino)-1-azabicyclo[2.2. 2]octane-3-acetonitrile] hydrochloride, talsaclidine, milameline, and xanomeline also produced dose-related decreases in avoidance responding. Our results demonstrate that muscarinic receptor agonists can decrease avoidance responding in a manner similar to dopamine-receptor antipsychotic drugs, suggesting that muscarinic receptor agonists may provide an alternative approach to the treatment of psychosis.


Subject(s)
Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Conditioning, Operant/drug effects , Dopamine Antagonists/pharmacology , Muscarinic Agonists/pharmacology , Animals , Cholinesterase Inhibitors/pharmacology , Depression, Chemical , Diazepam/pharmacology , Dose-Response Relationship, Drug , Haloperidol/pharmacology , Male , Pilocarpine/pharmacology , Rats , Rats, Inbred F344 , Receptors, Muscarinic/drug effects , Scopolamine/pharmacology , Succinimides/pharmacology
9.
J Med Chem ; 42(11): 1999-2006, 1999 Jun 03.
Article in English | MEDLINE | ID: mdl-10354408

ABSTRACT

Two new series of 1-(1,2,5-thiadiazol-4-yl)-4-azatricyclo[2.2.1.0(2, 6)]heptanes were synthesized and evaluated for their in vitro activity in cell lines transfected with either the human M1 or M2 receptor. 3-Phenyl-2-propyn-1-yloxy and -1-ylthio analogues substituted with halogen in the meta position showed high functional potency, efficacy, and selectivity toward the M1 receptor subtype. A quite unique functional M1 receptor selectivity was observed for compounds 8b, 8d, 8f, 9b, 9d, and 9f. Bioavailability studies in rats indicated an oral bioavailability of about 20-30%, with the N-oxide as the only detected metabolite.


Subject(s)
Aza Compounds/chemistry , Heptanes/chemistry , Muscarinic Agonists/chemistry , Receptors, Muscarinic/drug effects , Thiadiazoles/chemistry , Animals , Aza Compounds/chemical synthesis , Aza Compounds/pharmacokinetics , Aza Compounds/pharmacology , Binding, Competitive , Biological Availability , CHO Cells , Cell Line , Cerebral Cortex/metabolism , Cricetinae , Cyclic AMP/biosynthesis , Heptanes/chemical synthesis , Heptanes/pharmacokinetics , Heptanes/pharmacology , Humans , Hydrolysis , In Vitro Techniques , Mice , Muscarinic Agonists/chemical synthesis , Muscarinic Agonists/pharmacokinetics , Muscarinic Agonists/pharmacology , Phosphatidylinositols/metabolism , Radioligand Assay , Rats , Receptor, Muscarinic M1 , Receptor, Muscarinic M2 , Receptors, Muscarinic/metabolism , Structure-Activity Relationship , Thiadiazoles/chemical synthesis , Thiadiazoles/pharmacokinetics , Thiadiazoles/pharmacology , Transfection
10.
Life Sci ; 64(6-7): 527-34, 1999.
Article in English | MEDLINE | ID: mdl-10069519

ABSTRACT

The role of muscarinic receptors in schizophrenia was investigated using the muscarinic agonist PTAC. PTAC was highly selective for muscarinic receptors, was a partial agonist at muscarinic M2/M4 receptors and an antagonist at M1, M3 and M5 receptors. PTAC was highly active in animal models predictive of antipsychotic behavior including inhibition of conditioned avoidance responding in rats and blockade of apomorphine-induced climbing behavior in mice. d-Amphetamine-induced Fos expression in rat nucleus accumbens was inhibited by PTAC, thus directly demonstrating the ability of PTAC to modulate DA activity. In electrophysiological studies in rats, PTAC acutely inhibited the firing of A10 DA cells and after chronic administration decreased the number of spontaneously firing DA cells in the A10 brain area. However, PTAC did not appreciably alter the firing of A9 DA cells. Thus, PTAC appears to have novel antipsychotic-like activity and these data suggest that muscarinic compounds such as PTAC may represent a new class of antipsychotic agents.


Subject(s)
Antipsychotic Agents/pharmacology , Bridged Bicyclo Compounds/pharmacology , Receptors, Muscarinic/physiology , Schizophrenia/drug therapy , Thiadiazoles/pharmacology , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/metabolism , Antipsychotic Agents/therapeutic use , Behavior, Animal/drug effects , Binding, Competitive , Bridged Bicyclo Compounds/administration & dosage , Bridged Bicyclo Compounds/metabolism , Bridged Bicyclo Compounds/therapeutic use , CHO Cells , Catalepsy/chemically induced , Cricetinae , Dopamine/metabolism , Dopamine Agents/pharmacology , Dose-Response Relationship, Drug , Electrophysiology , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Neurons/drug effects , Neurons/physiology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Receptors, Muscarinic/metabolism , Schizophrenia/physiopathology , Second Messenger Systems/drug effects , Thiadiazoles/administration & dosage , Thiadiazoles/metabolism , Thiadiazoles/therapeutic use
11.
Neuroreport ; 9(15): 3481-6, 1998 Oct 26.
Article in English | MEDLINE | ID: mdl-9855303

ABSTRACT

(5R,6R) 6-(3-propylthio-1,2,5-thiadiazol-4-yl)-1-azabicyclo[3.2.1]oc tane (PTAC) is a selective muscarinic ligand with high affinity for central muscarinic receptors, agonist mode of action at the muscarinic M2 and M4 receptor subtypes and substantially less or no affinity for central dopamine receptors. In the present study PTAC, as well as the muscarinic agonists oxotremorine, RS86 and pilocarpine, inhibited dopamine D1 and D2 receptor agonist induced contralateral rotation in unilaterally 6-OHDA lesioned rats. The dose of SKF 38393 used to induce contralateral rotation also caused an intense Fos protein immunoreactivity in the rat dorsolateral striatum on the lesioned site which was inhibited by PTAC indicating that the inhibition of rotation by PTAC was not due to non-specific peripheral side effects.


Subject(s)
Cholinergic Fibers/drug effects , Dopamine Antagonists/pharmacology , Muscarinic Agonists/pharmacology , Oxotremorine/pharmacology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Antibodies , Antipsychotic Agents/pharmacology , Brain Chemistry/drug effects , Bridged Bicyclo Compounds/pharmacology , Cholinergic Fibers/chemistry , Dopamine/physiology , Dopamine Agonists/pharmacology , Male , Oxidopamine , Parasympathomimetics/pharmacology , Pilocarpine/pharmacology , Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-fos/immunology , Rats , Rats, Sprague-Dawley , Substantia Nigra/cytology , Substantia Nigra/metabolism , Succinimides/pharmacology , Sympatholytics , Thiadiazoles/pharmacology
12.
J Med Chem ; 41(22): 4378-84, 1998 Oct 22.
Article in English | MEDLINE | ID: mdl-9784113

ABSTRACT

Muscarinic agonists were tested in two models indicative of clinical antipsychotic activity: conditioned avoidance responding (CAR) in rats and inhibition of apomorphine-induced climbing in mice. The standard muscarinic agonists oxotremorine and pilocarpine were both active in these tests but showed little separation between efficacy and cholinergic side effects. Structure-activity relationships of the alkylthio-1,2,5-thiadiazole azacyclic type muscarinic partial agonists are shown, revealing the exo-6-(3-propyl/butylthio-1,2, 5-thiadiazol-4-yl)-1-azabicyclo[3.2.1]octane analogues (4a,b and 9a, b) to be the most potent antipsychotic agents with large separation between efficacy and cholinergic side effects. The lack of enantiomeric selectivity suggests the pharmacophoric elements are in the mirror plane of the compounds. A model explaining the potency differences of closely related compounds is offered. The data suggest that muscarinic agonists act as functional dopamine antagonists and that they could become a novel treatment of psychotic patients.


Subject(s)
Antipsychotic Agents/chemical synthesis , Dopamine Antagonists/chemical synthesis , Muscarinic Agonists/chemical synthesis , Thiadiazoles/chemical synthesis , Animals , Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacology , Antipsychotic Agents/toxicity , Avoidance Learning/drug effects , Brain/metabolism , Dopamine Antagonists/chemistry , Dopamine Antagonists/pharmacology , Dopamine Antagonists/toxicity , Drug Evaluation, Preclinical , In Vitro Techniques , Injections, Subcutaneous , Male , Mice , Models, Molecular , Molecular Conformation , Motor Activity/drug effects , Muscarinic Agonists/chemistry , Muscarinic Agonists/pharmacology , Muscarinic Agonists/toxicity , Rats , Rats, Sprague-Dawley , Salivation/drug effects , Stereoisomerism , Structure-Activity Relationship , Thiadiazoles/chemistry , Thiadiazoles/pharmacology , Thiadiazoles/toxicity , Tremor/chemically induced
13.
Eur J Pharmacol ; 356(2-3): 109-19, 1998 Sep 04.
Article in English | MEDLINE | ID: mdl-9774240

ABSTRACT

(5R,6R)6-(3-propylthio-1,2,5-thiadiazol-4-yl)-1-azabicyclo[3 .2.1]octane (PTAC) is a potent muscarinic receptor ligand with high affinity for central muscarinic receptors and no or substantially less affinity for a large number of other receptors or binding sites including dopamine receptors. The ligand exhibits partial agonist effects at muscarinic M2 and M4 receptors and antagonist effects at muscarinic M1, M3 and M5 receptors. PTAC inhibited conditioned avoidance responding, dopamine receptor agonist-induced behavior and D-amphetamine-induced FOS protein M5 expression in the nucleus accumbens without inducing catalepsy, tremor or salivation at pharmacologically relevant doses. The effect of PTAC on conditioned avoidance responding and dopamine receptor agonist-induced behavior was antagonized by the acetylcholine receptor antagonist scopolamine. The compound selectively inhibited dopamine cell firing (acute administration) as well as the number of spontaneously active dopamine cells (chronic administration) in the limbic ventral tegmental area (A10) relative to the non-limbic substantia nigra, pars compacta (A9). The results demonstrate that PTAC exhibits functional dopamine receptor antagonism despite its lack of affinity for the dopamine receptors and indicate that muscarinic receptor partial agonists may be an important new approach in the medical treatment of schizophrenia.


Subject(s)
Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Brain/drug effects , Bridged Bicyclo Compounds/pharmacology , Dopamine Antagonists/pharmacology , Motor Activity/drug effects , Receptors, Muscarinic/drug effects , Thiadiazoles/pharmacology , Animals , Antiparkinson Agents/pharmacology , Apomorphine/pharmacology , Brain/metabolism , Drug Interactions , Ligands , Male , Mice , Muscarinic Antagonists/pharmacology , Proto-Oncogene Proteins c-fos/drug effects , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Receptors, Muscarinic/metabolism , Scopolamine/pharmacology , Second Messenger Systems/drug effects
14.
Brain Res ; 795(1-2): 179-90, 1998 Jun 08.
Article in English | MEDLINE | ID: mdl-9622623

ABSTRACT

Activation of muscarinic m1 receptors which are coupled to the phosphoinositide (PI) second messenger transduction system is the initial objective of cholinergic replacement therapy in Alzheimer's disease. Thus, we evaluated the ability of the selective muscarinic receptor agonist (SMRA) xanomeline to stimulate in vivo phosphoinositide (PI) hydrolysis and compared it to a number of direct acting muscarinic agonists, two cholinesterase inhibitors and a putative m1 agonist/muscarinic m2 antagonist. Using a radiometric technique, it was determined that administration of xanomeline robustly stimulated in vivo PI hydrolysis and the effect was blocked by muscarinic antagonists, demonstrating mediation by muscarinic receptors. The non-selective muscarinic agonists pilocarpine, oxotremorine, RS-86, S-aceclidine, but not the less active isomer R-aceclidine, also effectively stimulated PI hydrolysis in mice. Amongst the putative m1 agonists, thiopilocarpine, hexylthio-TZTP as well as xanomeline effectively stimulated PI hydrolysis, but milameline, WAL 2014, SKB 202026 and PD 142505 did not significantly alter PI hydrolysis. Furthermore, WAL 2014 and SKB 202026 inhibited agonist-induced PI stimulation, suggesting that they act as antagonists at PI-coupled receptors in vivo. The cholinesterase inhibitors, tacrine and physostigmine, and the mixed muscarinic m1 agonist/m2 antagonist LU25-109 did not activate in vivo PI hydrolysis. Xanomeline, hexylthio-TZTP and thiopilocarpine were relatively free of cholinergic side effects, whereas milameline, WAL 2014 and SKB 202026 produced non-selective effects. Therefore, these data demonstrate that xanomeline selectively activates in vivo PI hydrolysis, consistent with activation of biochemical processes involved in memory and cognition and xanomeline's beneficial clinical effects on cognition in Alzheimers patients.


Subject(s)
Brain Chemistry/drug effects , Cholinergic Agents/metabolism , Muscarinic Agonists/pharmacology , Phosphatidylinositols/metabolism , Pyridines/pharmacology , Thiadiazoles/pharmacology , Animals , Binding, Competitive/physiology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cholinesterase Inhibitors/pharmacology , Dihydropyridines/pharmacology , Dose-Response Relationship, Drug , Hydrolysis , Hypothermia/chemically induced , Imines/pharmacology , Lithium/pharmacology , Male , Mice , Mice, Inbred Strains , Oximes/pharmacology , Parasympathomimetics/pharmacology , Physostigmine/pharmacology , Pilocarpine/pharmacology , Quinuclidines/pharmacology , Radioligand Assay , Salivation/drug effects , Tacrine/pharmacology , Tetrazoles/pharmacology , Tremor/chemically induced
15.
Biochem Biophys Res Commun ; 244(1): 156-60, 1998 Mar 06.
Article in English | MEDLINE | ID: mdl-9514902

ABSTRACT

Complementary DNA (cDNA) encoding human beta-amyloid precursor protein familial Alzheimer's disease (FAD) Swedish mutant (beta APPSM) form was cloned into a mammalian expression vector (PK255) containing the CMV promoter. The vector was transfected into Chinese hamster ovary cells containing human muscarinic m1 receptors (CHO-m1), and clonal cells stably expressing beta APPSM were isolated. The effects of m1-receptor activation by the selective m1 agonist xanomeline and the non-selective muscarinic agonist carbachol on processing of beta APPSM to release soluble APP (APPs) and beta-amyloid peptide (A beta) were compared. Xanomeline stimulated APP release with a potency 1000-fold greater than that observed for carbachol. Concentrations of carbachol and xanomeline producing maximal effects on APPs release reduced the secretion of A beta by 28 and 46%, respectively. These results extend previous studies with xanomeline and suggest that cholinergic replacement therapy for Alzheimer's disease may reduce amyloid deposition.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Muscarinic Agonists/pharmacology , Mutation , Protein Processing, Post-Translational/drug effects , Pyridines/pharmacology , Receptors, Muscarinic/physiology , Thiadiazoles/pharmacology , Transfection , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Blotting, Western , CHO Cells , Carbachol/pharmacology , Cloning, Molecular , Cricetinae , Humans , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Processing, Post-Translational/genetics , Receptor, Muscarinic M1 , Sweden
16.
J Med Chem ; 41(3): 379-92, 1998 Jan 29.
Article in English | MEDLINE | ID: mdl-9464368

ABSTRACT

The acetyl group of the muscarinic agonist aceclidine 4 was replaced by various 1,2,5-thiadiazoles to provide a new series of potent m1 muscarinic agonists 17 and 18. Optimal m1 muscarinic agonist potency was achieved when the 1,2,5-thiadiazole substituent was either a butyloxy, 17d, or butylthio, 18d, group. Although 1,2,5-oxadiazole 37 and pyrazine 39 are iso-pi-electronic with 1,2,5-thiadiazole 17d, both analogues were substantially less active than 17d. Compounds with high muscarinic affinity and/or m1 muscarinic agonist efficacy were also obtained when the 3-oxyquinuclidine moiety of 17d or 18c was replaced by ethanolamines, hydroxypyrrolidines, hydroxyazetidine, hydroxyisotropanes, or hydroxyazanorbornanes. The structure-activity data support the participation of the oxygen or sulfur atom in the substituent on the 1,2,5-thiadiazole in the activation of the m1 receptor. Several of these new 1,2,5-thiadiazoles have m1 agonist efficacy, potency, and selectivity comparable to those of xanomeline 2 in the muscarinic tests investigated.


Subject(s)
Muscarinic Agonists/chemistry , Muscarinic Agonists/pharmacology , Quinuclidines/chemistry , Quinuclidines/pharmacology , Thiadiazoles/chemistry , Animals , Brain/metabolism , Cell Line , Hydrolysis , Male , Mice , Models, Molecular , Muscarinic Agonists/metabolism , Phosphatidylinositols/metabolism , Quinuclidines/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley , Static Electricity
17.
J Med Chem ; 41(1): 109-16, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9438027

ABSTRACT

Conformationally constrained analogues of the potent muscarinic agonist 3-(4-methylthio)-1,2,5-thiadiazol-3-yl)-1,2,5,6-tetrahydro-1-methy lpyridine (methylthio-TZTP, 17) were designed and synthesized with the aim of (a) improving the antinociceptive selectivity over salivation and tremor and (b) predicting the active conformation of 17 with respect to the dihedral angle C4-C3-C3'-N2'. Using MOPAC 6.0 tricyclic analogues (7, 15, 16) with C4-C3-C3'-N2' dihedral angles close to 180 degrees and a rotation hindered analogue (9) with a C4-C3-C3'-N2' dihedral angle close to 274 degrees were designed, as these conformations had previously been suggested as being the active conformations. The analogues were tested for central muscarinic receptor binding affinity, for their antinociceptive activity in the mouse grid shock test, and, in the same assay, for their ability to induce tremor and salivation. The data showed that the tricyclic analogues (7, 15, 16) were equipotent with 17 as analgesics, but with no improved side effect profiles. The rotation-hindered analogue 9 had neither muscarinic receptor binding affinity nor antinociceptive activity. These results suggest that the active conformation of 17 has a C3-C4-C3'-N2' dihedral angle close to 180 degrees.


Subject(s)
Analgesics/chemical synthesis , Muscarinic Agonists/chemical synthesis , Pyridines/chemical synthesis , Receptors, Muscarinic/metabolism , Thiadiazoles/chemical synthesis , Analgesics/chemistry , Analgesics/metabolism , Analgesics/pharmacology , Animals , CHO Cells , Cell Membrane/metabolism , Cerebral Cortex/metabolism , Cricetinae , Electroshock , Male , Mice , Molecular Conformation , Molecular Structure , Muscarinic Agonists/chemistry , Muscarinic Agonists/metabolism , Muscarinic Agonists/pharmacology , Oxotremorine/metabolism , Pyridines/chemistry , Pyridines/metabolism , Pyridines/pharmacology , Rats , Rats, Wistar , Structure-Activity Relationship , Thiadiazoles/chemistry , Thiadiazoles/metabolism , Thiadiazoles/pharmacology
18.
Bioorg Med Chem Lett ; 8(20): 2897-902, 1998 Oct 20.
Article in English | MEDLINE | ID: mdl-9873644

ABSTRACT

Series of analogs to the functional m1 selective agonist, xanomeline (hexyloxy-TZTP), were evaluated for their in vitro m1 efficacy in cell lines transfected with the human m1 receptor. Systematic variation of the side chain and the azacyclic ring led to the discovery of potent muscarinic agonists with robust m1 efficacy, all having the phenylpropargyloxy/thio as the side chain. The most selective compound was the phenylpropargylthio-[3.2.1] endo analog 28, which is a potent and efficacious m1 agonist with no m2 activity.


Subject(s)
Muscarinic Agonists/chemistry , Pyridines/chemistry , Receptors, Muscarinic/metabolism , Thiadiazoles/chemistry , Animals , CHO Cells , Cells, Cultured , Cricetinae , Humans , Inhibitory Concentration 50 , Muscarinic Agonists/pharmacology , Pyridines/pharmacology , Rats , Receptor, Muscarinic M1 , Structure-Activity Relationship , Thiadiazoles/pharmacology , Transfection
19.
J Pharmacol Exp Ther ; 281(2): 868-75, 1997 May.
Article in English | MEDLINE | ID: mdl-9152396

ABSTRACT

The analgesic effects of a series of muscarinic agonists were investigated by use of the mouse acetic acid writhing, grid-shock, hot-plate and tail-flick tests. The compounds tested were oxotremorine, pilocarpine, arecoline, aceclidine, RS86 and four 3-3(substituted-1,2,5-thiadiazol-4-yl)-1,2,5,6-tetrahy-dro-1 -methyl pyridines (substituted TZTP), these being propoxy-TZTP, 3-Cl-propylthio-TZTP, xanomeline (hexyloxy-TZTP) and hexylthio-TZTP. These agonists were also assayed for their ability to displace [3H]oxotremorine-M and [3H]pirenz-epine binding and for their functional selectivity at pharmaco-logic M1, M2 and M3 receptors. These compounds all produced dose-dependent antinociceptive effects in all of the mouse analgesia tests. The effects of oxotremorine in the writhing test were fully antagonized by the muscarinic antagonist scopolamine (0.1 mg/kg), but only partially antagonized by methsco-polamine (10 mg/kg) and unaffected by the opioid antagonist naltrexone. 3-Cl-propylthio-TZTP and propoxy-TZTP had virtually no effect at the M1 receptor subtype as measured by the human m1 clone expressed in baby hamster kidney cells or the rabbit vas deferens assay. These compounds, however, were more potent in the analgesia tests than the selective M1 agonists xanomeline and hexylthio-TZTP. These data suggest that muscarinic analgesia is mediated by central muscarinic receptors. However, activity at the M1 receptor subtype is not a requirement for antinociceptive activity.


Subject(s)
Analgesics/pharmacology , Muscarinic Agonists/pharmacology , Receptors, Muscarinic/drug effects , Animals , Atrial Function , Cell Line , Cricetinae , Guinea Pigs , Heart Atria/drug effects , Humans , Hydrolysis , In Vitro Techniques , Inositol Phosphates/metabolism , Male , Mice , Muscle Contraction/drug effects , N-Methylscopolamine , Naltrexone/pharmacology , Rabbits , Rats , Rats, Wistar , Receptor, Muscarinic M1 , Scopolamine/pharmacology , Scopolamine Derivatives/pharmacology , Transfection , Vas Deferens/drug effects , Vas Deferens/physiology
20.
J Pharmacol Exp Ther ; 281(2): 876-83, 1997 May.
Article in English | MEDLINE | ID: mdl-9152397

ABSTRACT

Butylthio[2.2.2] ((+)-(S)-3-(4-(Butylthio)-1,2,5-thiadiazol-3-yl)-1-azabicyclo[2.2. 2] octane) is an agonist/antagonist at muscarinic receptors. The analgesic potential of butylthio[2.2.2] was assessed in the mouse by use of the grid-shock, tail-flick, hotplate and writhing tests. The ED50 values ranged from 0.19 to 1.47 mg/kg and 1.51 to 12.23 mg/kg 30 min after s.c. and p.o. administration, respectively, yielding p.o./s.c. ratios ranging from 7 to 27. The ED50 values for salivation and tremor were > 30 and 12.31 mg/kg s.c., and > 60 and > 60 mg/kg p.o., yielding therapeutic windows > 130 and 54, and, > 40 and > 40, after s.c. and p.o. administration, respectively. Motor impairment or lethality were only seen at doses 116 and 254 times higher than the antinociceptive doses. Butylthio[2.2.2] was equieffective to, and 3- to 24-fold more potent than morphine. The duration of action was similar to that of morphine. The dose-response curve was shifted dose dependently to the right by the muscarinic antagonist scopolamine but not by the opioid antagonist naltrexone. The antinociceptive effect of butylthio[2.2.2] was reversed by the centrally acting muscarinic antagonist scopolamine but not by the peripherally acting muscarinic antagonist methscopolamine. After 6.5 days repeated dosing in mice, morphine produced marked tolerance, whereas butylthio[2.2.2] produced minimal, if any, tolerance. In the rat grid-shock test, ED50 values of 0.26 mg/kg s.c. and 25.28 mg/kg p.o. were obtained. These data show that butylthio[2.2.2] is a potent and efficacious antinociceptive with a very favorable therapeutic window after s.c. and p.o. administration in mice, and with good efficacy in rats.


Subject(s)
Analgesics/pharmacology , Cholinergic Agents/pharmacology , Thiadiazoles/pharmacology , Administration, Oral , Animals , Cholinergic Agents/administration & dosage , Dose-Response Relationship, Drug , Male , Mice , Morphine/pharmacology , Rats , Thiadiazoles/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...