Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Psychopharmacology (Berl) ; 232(21-22): 4033-41, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26177580

ABSTRACT

The low rate of success for identifying effective treatments for cognitive dysfunction has prompted recent efforts to improve pharmaceutical discovery and development. In particular, investigators have emphasized improving translation from pre-clinical to clinical research. A specific area of focus has been touchscreen technology; this computer-automated behavioral testing method provides an objective assessment of performance that can be used across species. As part of a larger multi-site study with partners from the Innovative Medicines Initiative (IMI), two US sites, AbbVie and Pfizer, conducted a cross-site experiment with a common protocol for the visual discrimination (VD) task using identical testing equipment, stimuli, and rats of the same strains, sex, and age from the same supplier. As most touchscreen-based rodent experiments have used Lister-Hooded rats that are not readily available outside of Europe, a strain comparison with male Long-Evans rats was conducted as part of the study. Rats were trained for asymptotic performance, and test sessions were performed once per week in a full crossover design with cognition-impairing drugs. Drugs tested were phencyclidine and S-ketamine (N-methyl-D-aspartate (NMDA) antagonists), D-amphetamine (indirect dopamine agonist), and scopolamine (muscarinic antagonist). Satellite brain and plasma samples were taken to confirm appropriate exposures. Results indicate that both rat strains show similar patterns of impairment, although Lister-Hooded rats were more sensitive than Long-Evans rats to three out of four drugs tested. This suggests that researchers should fully explore dose-response relationships in their strain of choice and use care in the interpretation of reversal of cognitive impairment.


Subject(s)
Cognition/drug effects , Dextroamphetamine/pharmacology , Discrimination Learning/drug effects , Ketamine/pharmacology , Phencyclidine/pharmacology , Scopolamine/pharmacology , Visual Perception/drug effects , Animals , Choice Behavior/drug effects , Dopamine Agonists/pharmacology , Europe , Excitatory Amino Acid Antagonists/pharmacology , Male , Muscarinic Antagonists/pharmacology , Rats , Rats, Long-Evans
2.
J Neurosci ; 34(32): 10592-602, 2014 Aug 06.
Article in English | MEDLINE | ID: mdl-25100593

ABSTRACT

The elevation of kynurenic acid (KYNA) observed in schizophrenic patients may contribute to core symptoms arising from glutamate hypofunction, including cognitive impairments. Although increased KYNA levels reduce excitatory neurotransmission, KYNA has been proposed to act as an endogenous antagonist at the glycine site of the glutamate NMDA receptor (NMDAR) and as a negative allosteric modulator at the α7 nicotinic acetylcholine receptor. Levels of KYNA are elevated in CSF and the postmortem brain of schizophrenia patients, and these elevated levels of KYNA could contribute to NMDAR hypofunction and the cognitive deficits and negative symptoms associated with this disease. However, the impact of endogenously produced KYNA on brain function and behavior is less well understood due to a paucity of pharmacological tools. To address this issue, we identified PF-04859989, a brain-penetrable inhibitor of kynurenine aminotransferase II (KAT II), the enzyme responsible for most brain KYNA synthesis. In rats, systemic administration of PF-04859989 dose-dependently reduced brain KYNA to as little as 28% of basal levels, and prevented amphetamine- and ketamine-induced disruption of auditory gating and improved performance in a sustained attention task. It also prevented ketamine-induced disruption of performance in a working memory task and a spatial memory task in rodents and nonhuman primates, respectively. Together, these findings support the hypotheses that endogenous KYNA impacts cognitive function and that inhibition of KAT II, and consequent lowering of endogenous brain KYNA levels, improves cognitive performance under conditions considered relevant for schizophrenia.


Subject(s)
Brain/metabolism , Cognition/physiology , Kynurenic Acid/metabolism , Schizophrenia/cerebrospinal fluid , Schizophrenia/pathology , Animals , Attention/drug effects , Attention/physiology , Enzyme Inhibitors/pharmacology , Evoked Potentials, Auditory/drug effects , Evoked Potentials, Auditory/physiology , Female , Hippocampus/cytology , Humans , Macaca mulatta , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Neurons/drug effects , Neurons/physiology , Pyrazoles/pharmacology , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Wakefulness
3.
J Pharmacol Exp Ther ; 341(2): 396-409, 2012 May.
Article in English | MEDLINE | ID: mdl-22328573

ABSTRACT

Cyclic nucleotides are critical regulators of synaptic plasticity and participate in requisite signaling cascades implicated across multiple neurotransmitter systems. Phosphodiesterase 9A (PDE9A) is a high-affinity, cGMP-specific enzyme widely expressed in the rodent central nervous system. In the current study, we observed neuronal staining with antibodies raised against PDE9A protein in human cortex, cerebellum, and subiculum. We have also developed several potent, selective, and brain-penetrant PDE9A inhibitors and used them to probe the function of PDE9A in vivo. Administration of these compounds to animals led to dose-dependent accumulation of cGMP in brain tissue and cerebrospinal fluid, producing a range of biological effects that implied functional significance for PDE9A-regulated cGMP in dopaminergic, cholinergic, and serotonergic neurotransmission and were consistent with the widespread distribution of PDE9A. In vivo effects of PDE9A inhibition included reversal of the respective disruptions of working memory by ketamine, episodic and spatial memory by scopolamine, and auditory gating by amphetamine, as well as potentiation of risperidone-induced improvements in sensorimotor gating and reversal of the stereotypic scratching response to the hallucinogenic 5-hydroxytryptamine 2A agonist mescaline. The results suggested a role for PDE9A in the regulation of monoaminergic circuitry associated with sensory processing and memory. Thus, PDE9A activity regulates neuronal cGMP signaling downstream of multiple neurotransmitter systems, and inhibition of PDE9A may provide therapeutic benefits in psychiatric and neurodegenerative diseases promoted by the dysfunction of these diverse neurotransmitter systems.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Cholinergic Agents/pharmacology , Cyclic GMP/metabolism , Neurons/drug effects , Neurons/metabolism , Phosphodiesterase Inhibitors/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Avoidance Learning/drug effects , Brain/drug effects , Brain/metabolism , Female , Humans , Macaca fascicularis , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neurotransmitter Agents/pharmacology , Rats , Rats, Long-Evans , Rats, Wistar , Sensory Gating/drug effects , Stereotyped Behavior/drug effects , Synaptic Transmission/drug effects
4.
J Med Chem ; 54(13): 4536-47, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21650160

ABSTRACT

Utilizing structure-based virtual library design and scoring, a novel chimeric series of phosphodiesterase 10A (PDE10A) inhibitors was discovered by synergizing binding site interactions and ADME properties of two chemotypes. Virtual libraries were docked and scored for potential binding ability, followed by visual inspection to prioritize analogs for parallel and directed synthesis. The process yielded highly potent and selective compounds such as 16. New X-ray cocrystal structures enabled rational design of substituents that resulted in the successful optimization of physical properties to produce in vivo activity and to modulate microsomal clearance and permeability.


Subject(s)
Antipsychotic Agents/chemical synthesis , Phosphodiesterase Inhibitors/chemical synthesis , Phosphoric Diester Hydrolases/metabolism , Schizophrenia/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antipsychotic Agents/pharmacokinetics , Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Binding Sites , Blood-Brain Barrier/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Crystallography, X-Ray , Cyclic GMP/metabolism , Databases, Factual , Drug Design , Humans , In Vitro Techniques , Mice , Mice, Knockout , Microsomes, Liver/metabolism , Models, Molecular , Permeability , Phosphodiesterase Inhibitors/pharmacokinetics , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/genetics , Protein Conformation , Structure-Activity Relationship
5.
J Med Chem ; 52(16): 5188-96, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19630403

ABSTRACT

By utilizing structure-based drug design (SBDD) knowledge, a novel class of phosphodiesterase (PDE) 10A inhibitors was identified. The structure-based drug design efforts identified a unique "selectivity pocket" for PDE10A inhibitors, and interactions within this pocket allowed the design of highly selective and potent PDE10A inhibitors. Further optimization of brain penetration and drug-like properties led to the discovery of 2-[4-(1-methyl-4-pyridin-4-yl-1H-pyrazol-3-yl)-phenoxymethyl]-quinoline (PF-2545920). This PDE10A inhibitor is the first reported clinical entry for this mechanism in the treatment of schizophrenia.


Subject(s)
Antipsychotic Agents/chemical synthesis , Models, Molecular , Phosphoric Diester Hydrolases/metabolism , Pyrazoles/chemical synthesis , Quinolines/chemical synthesis , Schizophrenia/drug therapy , Animals , Antipsychotic Agents/pharmacokinetics , Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Binding Sites , Brain/metabolism , Crystallography, X-Ray , Dogs , Female , Humans , Hydrogen Bonding , In Vitro Techniques , Macaca fascicularis , Male , Mice , Mice, Knockout , Microsomes, Liver/metabolism , Molecular Structure , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/genetics , Protein Binding , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Quinolines/pharmacokinetics , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
6.
Psychopharmacology (Berl) ; 197(1): 115-26, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18060387

ABSTRACT

RATIONALE: Phosphodiesterases (PDEs) belonging to the PDE4 family control intracellular concentrations of cyclic adenosine monophosphate (cAMP) by catalyzing its hydrolysis. Four separate PDE4 genes (PDE4A, PDE4B, PDE4C, and PDE4D) have been identified. PDE4 has been reported to be involved in various central nervous system (CNS) functions including depression, memory, and schizophrenia, although the specific subtype mediating these effects remains unclear. OBJECTIVE: To investigate the role of PDE4B in the CNS, PDE4B wild-type and knockout mice (C57BL/6N background) were assessed in a variety of well-characterized behavioral tasks, and their brains were assayed for monoamine content. RESULTS: Knockout mice showed a significant reduction in prepulse inhibition. Spontaneous locomotor activity was decreased (16%) in knockout mice. Furthermore, when challenged with amphetamine, both groups of mice responded similarly to a low dose of d-amphetamine (1.0 mg/kg), but knockout mice showed an enhanced response to a higher dose (1.78 mg/kg). Decreases in baseline levels of monoamines and their metabolites within the striatum of knockout mice were also observed. PDE4B knockout mice showed a modest decrease in immobility time in the forced swim test that approached significance. In several other tests, including the elevated plus maze, hot plate, passive avoidance, and Morris water maze, wild-type and knockout mice performed similarly. CONCLUSION: The present studies demonstrate decreased striatal DA and 5-HT activity in the PDE4B knockout mice associated with decreased prepulse inhibition, decreased baseline motor activity, and an exaggerated locomotor response to amphetamine. These data further support a role for PDE4B in psychiatric diseases and striatal function.


Subject(s)
Behavior, Animal/physiology , Brain/physiopathology , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Dopamine/metabolism , Serotonin/metabolism , Animals , Arousal/drug effects , Arousal/physiology , Behavior, Animal/drug effects , Brain/drug effects , Dextroamphetamine/pharmacology , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motivation , Motor Activity/drug effects , Motor Activity/physiology , Reflex, Startle/drug effects , Reflex, Startle/physiology
7.
Neuropharmacology ; 54(2): 417-27, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18061215

ABSTRACT

The phenotype of genetically modified animals is strongly influenced by both the genetic background of the animal as well as environmental factors. We have previously reported the behavioral and neurochemical characterization of PDE10A knockout mice maintained on a DBA1LacJ (PDE10A(DBA)) genetic background. The aim of the present studies was to assess the behavioral and neurochemical phenotype of PDE10A knockout mice on an alternative congenic C57BL/6N (PDE10A(C57)) genetic background. Consistent with our previous results, PDE10A(C57) knockout mice showed a decrease in exploratory locomotor activity and a delay in the acquisition of conditioned avoidance responding. Also consistent with previous studies, the elimination of PDE10A did not alter basal levels of striatal cGMP or cAMP or affect behavior in several other well-characterized behavioral assays. PDE10A(C57) knockout mice showed a blunted response to MK-801, although to a lesser degree than previously observed in the PDE10A(DBA) knockout mice, and no differences were observed following a PCP challenge. PDE10A(C57) knockout mice showed a significant change in striatal dopamine turnover, which was accompanied by an enhanced locomotor response to AMPH, These studies demonstrate that while many of the behavioral effects of the PDE10A gene deletion appear to be independent of genetic background, the impact of the deletion on behavior can vary in magnitude. Furthermore, the effects on the dopaminergic system appear to be background-dependent, with significant effects observed only in knockout mice on the C57BL6N genetic background.


Subject(s)
Behavior, Animal/physiology , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/physiology , Amphetamine/pharmacology , Animals , Anxiety/psychology , Avoidance Learning/drug effects , Avoidance Learning/physiology , Behavior, Animal/drug effects , Biogenic Monoamines/metabolism , Brain Chemistry/drug effects , Brain Chemistry/genetics , Chromatography, High Pressure Liquid , Depression/psychology , Dizocilpine Maleate/pharmacology , Dopamine/metabolism , Dopamine Uptake Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hot Temperature , Methamphetamine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Nucleotides, Cyclic/metabolism , Pain Measurement/drug effects , Phencyclidine/pharmacology , Phosphoproteins/metabolism , Serotonin/metabolism , Swimming/psychology
8.
Psychopharmacology (Berl) ; 192(3): 415-24, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17333137

ABSTRACT

RATIONALE: Recent studies provide evidence for reduced phosphodiesterase-4B (PDE4B) as a genetic susceptibility factor as well as suggesting an association of several single nucleotide polymorphisms (SNPs) in PDE4B that are associated with an increased incidence of schizophrenia. OBJECTIVES: The aim of the current study was to assess the activity of rolipram, a nonsubtype-selective PDE4 inhibitor, in several animal models predictive of antipsychotic-like efficacy and side-effect liability and to use PDE4B wild-type and knockout mice to begin to understand the subtypes involved in the activity of rolipram. RESULTS: In rats, rolipram antagonized both phencyclidine hydrochloride- and D-amphetamine-induced hyperactivity and inhibited conditioned avoidance responding (CAR). In PDE4B wild-type mice, rolipram dose-dependently suppressed CAR (ED(50) = 2.4 mg/kg); however, in knockout mice, their sensitivity to rolipram at the higher doses (1.0 and 3.2 mg/kg) was reduced, resulting in a threefold shift in the ED(50) (7.3 mg/kg), suggesting PDE4B is involved, at least in part, with the activity of rolipram. Only the highest dose of rolipram (3.2 mg/kg) produced a modest but significant degree of catalepsy. CONCLUSIONS: Rolipram has a pharmacologic profile similar to that of the atypical antipsychotics and has low extrapyramidal symptom liability. These results suggest that PDE4B mediates the antipsychotic effects of rolipram in CAR and that the PDE4B-regulated cyclic adenosine monophosphate signaling pathway may play a role in the pathophysiology and pharmacotherapy of psychosis.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/genetics , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Rolipram/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/adverse effects , Avoidance Learning/drug effects , Catalepsy/chemically induced , Conditioning, Operant/drug effects , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4 , Disease Models, Animal , Dose-Response Relationship, Drug , Hyperkinesis/chemically induced , Hyperkinesis/drug therapy , Male , Mice , Mice, Inbred DBA , Mice, Knockout , Motor Activity/drug effects , Polymorphism, Genetic , Psychotic Disorders/drug therapy , Psychotic Disorders/physiopathology , Rats , Rolipram/administration & dosage , Rolipram/adverse effects , Schizophrenia/drug therapy , Schizophrenia/physiopathology , Signal Transduction
9.
Neuropharmacology ; 52(2): 279-90, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16949622

ABSTRACT

CP-809,101 is a potent, functionally selective 5-HT(2C) agonist that displays approximately 100% efficacy in vitro. The aim of the present studies was to assess the efficacy of a selective 5-HT(2C) agonist in animal models predictive of antipsychotic-like efficacy and side-effect liability. Similar to currently available antipsychotic drugs, CP-809,101 dose-dependently inhibited conditioned avoidance responding (CAR, ED(50)=4.8 mg/kg, sc). The efficacy of CP-809,101 in CAR was completely antagonized by the concurrent administration of the 5-HT(2C) receptor antagonist, SB-224,282. CP-809,101 antagonized both PCP- and d-amphetamine-induced hyperactivity with ED(50) values of 2.4 and 2.9 mg/kg (sc), respectively and also reversed an apomorphine induced-deficit in prepulse inhibition. At doses up to 56 mg/kg, CP-809,101 did not produce catalepsy. Thus, the present results demonstrate that the 5-HT(2C) agonist, CP-809,101, has a pharmacological profile similar to that of the atypical antipsychotics with low extrapyramidal symptom liability. CP-809,101 was inactive in two animal models of antidepressant-like activity, the forced swim test and learned helplessness. However, CP-809,101 was active in novel object recognition, an animal model of cognitive function. These data suggest that 5-HT(2C) agonists may be a novel approach in the treatment of psychosis as well as for the improvement of cognitive dysfunction associated with schizophrenia.


Subject(s)
Antipsychotic Agents/therapeutic use , Psychotic Disorders/drug therapy , Serotonin 5-HT2 Receptor Agonists , Serotonin Receptor Agonists/therapeutic use , Amphetamines , Animals , Antipsychotic Agents/chemistry , Avoidance Learning/drug effects , Behavior, Animal , Catalepsy/chemically induced , Catalepsy/drug therapy , Dextroamphetamine , Disease Models, Animal , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Helplessness, Learned , Humans , Hyperkinesis/chemically induced , Hyperkinesis/drug therapy , Inhibition, Psychological , Male , Mice , Motor Activity/drug effects , NIH 3T3 Cells , Piperazines/chemistry , Piperazines/therapeutic use , Protein Binding/drug effects , Psychotic Disorders/etiology , Psychotic Disorders/physiopathology , Pyrazines/chemistry , Pyrazines/therapeutic use , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT2C/physiology
10.
Neuropharmacology ; 51(2): 386-96, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16780899

ABSTRACT

Phosphodiesterase 10A (PDE10A) is a recently identified cyclic nucleotide phosphodiesterase expressed primarily in dopaminoreceptive medium spiny neurons of the striatum. We report that papaverine is a potent, specific inhibitor of PDE10A and use this compound to explore the role of PDE10A in regulating striatal function. Papaverine administration produces an increase in striatal tissue levels of cGMP and an increase in extracellular cAMP measured by microdialysis. These cyclic nucleotide changes are accompanied by increases in the phosphorylation of CREB and ERK, downstream markers of neuronal activation. In rats, papaverine potentiates haloperidol-induced catalepsy, consistent with the hypothesis that inhibition of PDE10A can increase striatal output and prompting a further evaluation of papaverine in models predictive of antipsychotic activity. Papaverine is found to inhibit conditioned avoidance responding in rats and mice and to inhibit PCP- and amphetamine-stimulated locomotor activity in rats. The effects of papaverine on striatal cGMP and CREB and ERK phosphorylation, as well as on conditioned avoidance responding, were absent in PDE10A knockout mice, indicating that the effects of the compound are the result of PDE10A inhibition. These results indicate that PDE10A regulates the activation of striatal medium spiny neurons through effects on cAMP- and cGMP-dependent signaling cascades. Furthermore, the present results demonstrate that papaverine has efficacy in behavioral models predictive of antipsychotic activity. Thus, inhibition of PDE10A may represent a novel approach to the treatment of psychosis.


Subject(s)
Antipsychotic Agents/pharmacology , Corpus Striatum/enzymology , Papaverine/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/physiology , Animals , Avoidance Learning/drug effects , Catalepsy/chemically induced , Central Nervous System Stimulants/pharmacology , Corpus Striatum/metabolism , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic GMP/metabolism , Dendritic Spines/drug effects , Dendritic Spines/physiology , Dextroamphetamine/pharmacology , Drug Synergism , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Haloperidol/pharmacology , Mice , Mice, Knockout , Motor Activity/drug effects , Phencyclidine/pharmacology , Phosphorylation , Rats
11.
Neuropharmacology ; 51(2): 374-85, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16769090

ABSTRACT

PDE10A is a newly identified phosphodiesterase that is highly expressed by the medium spiny projection neurons of the striatum. In order to investigate the physiological role of PDE10A in the central nervous system, PDE10A knockout mice (PDE10A(-/-)) were characterized both behaviorally and neurochemically. PDE10A(-/-) mice showed decreased exploratory activity and a significant delay in the acquisition of conditioned avoidance behavior when compared to wild-type (PDE10A(+/+)) mice. However, in a variety of other well-characterized behavioral tasks, including the elevated plus maze (anxiety), forced swim test (depression), hot plate (nociception) and two memory models (passive avoidance and Morris water maze), PDE10A(-/-) mice performed similarly to wild-type mice. When challenged with PCP or MK-801, PDE10A(-/-) mice showed a blunted locomotor response in comparison to PDE10A(+/+) mice. In contrast, PDE10A(-/-) and PDE10A(+/+) mice responded similarly to the locomotor stimulating effects of amphetamine and methamphetamine. Our findings suggest that PDE10A is involved in regulating striatal output, possibly by reducing the sensitivity of medium spiny neurons to glutamatergic excitation. These results are discussed in relationship to the hypothesis that PDE10A inhibition presents a novel treatment for psychosis.


Subject(s)
Corpus Striatum/physiology , Phosphoric Diester Hydrolases/physiology , Amphetamine/pharmacology , Animals , Avoidance Learning , Behavior, Animal , Biogenic Monoamines/metabolism , Brain/metabolism , Central Nervous System Stimulants/pharmacology , Conditioning, Operant , Corpus Striatum/enzymology , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Maze Learning , Methamphetamine/pharmacology , Mice , Mice, Knockout , Motor Activity/drug effects , Phencyclidine/pharmacology , Phosphoric Diester Hydrolases/genetics
12.
Eur J Pharmacol ; 486(1): 9-17, 2004 Feb 13.
Article in English | MEDLINE | ID: mdl-14751402

ABSTRACT

Cholinesterase inhibitors vary in their selectivity for acetylcholinesterase versus butyrylcholinesterase. We examined several cholinesterase inhibitors and assessed the relative role of acetylcholinesterase versus butyrylcholinesterase inhibition in central and peripheral responses to these medications. Donepezil and icopezil are highly selective for acetylcholinesterase, whereas tacrine and heptylphysostigmine demonstrated greater potency for butyrylcholinesterase over acetylcholinesterase. All four compounds increased acetylcholine levels in mouse brains. Dose-response curves for tremor (central effect) and salivation (peripheral effect) showed that donepezil and icopezil possess a more favourable therapeutic index than the nonselective inhibitors, tacrine and heptylphysostigmine. Co-administration of the selective butyrylcholinesterase inhibitor tetraisopropylpyrophosphoramide (iso-OMPA) potentiated peripheral, but not central, effects of the selective acetylcholinesterase inhibitor icopezil. The improved therapeutic index observed in mice with icopezil is due to a high degree of selectivity for acetylcholinesterase versus butyrylcholinesterase, suggesting that high selectivity for acetylcholinesterase may contribute to the clinically favourable tolerability profile of agents such as donepezil in Alzheimer's disease patients.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Cholinesterase Inhibitors/pharmacology , Physostigmine/analogs & derivatives , Acetylcholine/metabolism , Animals , Brain/drug effects , Brain/metabolism , Butyrylcholinesterase/blood , Cholinesterase Inhibitors/therapeutic use , Donepezil , Dose-Response Relationship, Drug , Humans , Indans/pharmacology , Male , Mice , Physostigmine/pharmacology , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley , Tacrine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...