Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
1.
Annu Rev Pharmacol Toxicol ; 57: 171-187, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27575715

ABSTRACT

There are many challenges along the path to the approval of new drugs to treat CNS disorders, one of the greatest areas of unmet medical need with a large societal burden and health-care impact. Unfortunately, over the past two decades, few CNS drug approvals have succeeded, leading many pharmaceutical companies to deprioritize this therapeutic area. The reasons for the failures in CNS drug discovery are likely to be multifactorial. However, selecting the most biologically plausible molecular targets that are relevant to the disorder is a critical first step to improve the probability of success. In this review, we outline previous methods for identifying and validating novel targets for CNS drug discovery, and, cognizant of previous failures, we discuss potential new strategies that may improve the probability of success of developing novel treatments for CNS disorders.


Subject(s)
Central Nervous System Agents/administration & dosage , Central Nervous System Diseases/drug therapy , Drug Discovery/methods , Drug Discovery/standards , Mental Disorders/drug therapy , Animals , Central Nervous System Agents/metabolism , Central Nervous System Diseases/genetics , Central Nervous System Diseases/metabolism , Humans , Mental Disorders/genetics , Mental Disorders/metabolism , Models, Animal , Reproducibility of Results
2.
Neuropharmacology ; 61(4): 665-76, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21619887

ABSTRACT

Inhibition of phosphodiesterase 9 (PDE9) has been reported to enhance rodent cognitive function and may represent a potential novel approach to improving cognitive dysfunction in Alzheimer's disease. PF-04447943, (6-[(3S,4S)-4-methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-1-(tetrahydro-2H-pyran-4-yl)-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one), a recently described PDE9 inhibitor, was found to have high affinity (Ki of 2.8, 4.5 and 18 nM) for human, rhesus and rat recombinant PDE9 respectively and high selectivity for PDE9 versus PDEs1-8 and 10-11. PF-04447943 significantly increased neurite outgrowth and synapse formation (as indicated by increased synapsin 1 expression) in cultured hippocampal neurons at low (30-100 nM) but not high (300-1000 nM) concentrations. PF-04447943 significantly facilitated hippocampal slice LTP evoked by a weak tetanic stimulus at a concentration of 100 nM but failed to affect response to the weak tetanus at either 30 or 300 nM, or the LTP produced by a theta burst stimulus. Systemic administration of PF-04447943 (1-30 mg/kg p.o.) dose-dependently increased cGMP in the cerebrospinal fluid 30 min after administration indicating target engagement in the CNS of rats. PF-04447943 (1-3 mg/kg p.o.) significantly improved cognitive performance in three rodent cognition assays (mouse Y maze spatial recognition memory model of natural forgetting, mouse social recognition memory model of natural forgetting and rat novel object recognition with a scopolamine deficit). When administered at a dose of 3 mg/kg p.o., which improved performance in novel object recognition, PF-04447943 significantly increased phosphorylated but not total GluR1 expression in rat hippocampal membranes. Collectively these data indicate that PF-04447943 is a potent, selective brain penetrant PDE9 inhibitor that increased indicators of hippocampal synaptic plasticity and improved cognitive function in a variety of cognition models in both rats and mice. Results with PF-04447943 are consistent with previously published findings using a structurally diverse PDE9 inhibitor, BAY73-6199, and further support the suggestion that PDE9 inhibition may represent a novel approach to the palliative remediation of cognitive dysfunction.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Cognition/drug effects , Neuronal Plasticity/drug effects , Phosphodiesterase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidinones/pharmacology , Synapses/drug effects , Synapses/enzymology , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , CHO Cells , Cognition/physiology , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Female , HEK293 Cells , Hippocampus/drug effects , Hippocampus/enzymology , Humans , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neuronal Plasticity/physiology , Phosphodiesterase Inhibitors/metabolism , Pyrazoles/metabolism , Pyrimidinones/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar
3.
J Psychopharmacol ; 23(5): 475-86, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19395429

ABSTRACT

Caffeine is known to disrupt sleep and its administration to human subjects has been used to model sleep disruption. We previously showed that its effects on sleep onset latency are comparable between rats and humans. This study evaluated the potential use of caffeine as a model of sleep disruption in the rat, by assessing its effects on sleep architecture and electroencephalogram (EEG) frequency spectrum, and using sleep-promoting drugs to reverse these effects. Rats were implanted with radiotelemetry devices for body temperature, EEG, electromyogram and locomotor activity. Following recovery, animals were dosed with caffeine (10 mg/kg) alone or in combination with zolpidem (10 mg/kg) or trazodone (20 mg/kg). Sleep was scored for the subsequent 12 h using automated analysis software. Caffeine dose-dependently disrupted sleep: it increased WAKE time, decreased NREM (non-REM) sleep time and NREM bout duration (but not bout number), and decreased delta activity in NREM sleep. It also dose-dependently increased locomotor activity and body temperature. When given alone, zolpidem suppressed REM whilst trazodone increased NREM sleep time at the expense of WAKE, increased NREM bout duration, increased delta activity in NREM sleep and reduced body temperature. In combination, zolpidem attenuated caffeine's effects on WAKE, whilst trazodone attenuated its effects on NREM sleep, NREM bout duration, delta activity, body temperature and locomotor activity. Caffeine administration produced many of the signs of insomnia that were improved by two of its most successful current treatments. This model may therefore be useful in the study of new drugs for the treatment of sleep disturbance.


Subject(s)
Caffeine/toxicity , Central Nervous System Stimulants/toxicity , Sleep Initiation and Maintenance Disorders/chemically induced , Sleep/drug effects , Animals , Body Temperature/drug effects , Disease Models, Animal , Electroencephalography , Electromyography , Hypnotics and Sedatives/pharmacology , Male , Motor Activity/drug effects , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Sleep Initiation and Maintenance Disorders/drug therapy , Telemetry , Trazodone/pharmacology , Zolpidem
4.
Neurochem Res ; 34(10): 1847-56, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19399611

ABSTRACT

Mutations in Cu/Zn superoxide dismutase are a cause of motor neuron death in about 20% of cases of familial amyotrophic lateral sclerosis (ALS). Although the molecular mechanism of which these mutations induce motor neuron cell death is to a large extent unknown, there is significant evidence that effects on mitochondrial function and development of oxidative stress make a major contribution to the selective death of motor neurons in this disease. In this overview article we review the current understanding of mutant SOD1-mediated motor neuron degeneration in ALS with focus on oxidative damage and mitochondrial dysfunction. We also present novel information on the role of mitochondrial glutathione for the survival of NSC-34 cells stably transfected with the human SOD1(G93A) mutation, putting forward the hypothesis that this antioxidant pool provides a potentially useful target for therapeutic intervention.


Subject(s)
Amino Acid Substitution/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Apoptosis/genetics , Glutathione/genetics , Mitochondria/genetics , Motor Neurons/pathology , Superoxide Dismutase/genetics , Alanine/genetics , Amyotrophic Lateral Sclerosis/enzymology , Animals , Cell Death/genetics , Glutathione/metabolism , Glycine/genetics , Humans , Mitochondria/metabolism , Mitochondria/pathology , Motor Neurons/enzymology , Oxidative Stress/genetics , Superoxide Dismutase/metabolism
5.
J Psychopharmacol ; 23(5): 487-94, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19351801

ABSTRACT

Caffeine is the world's most popular stimulant and is known to disrupt sleep. Administration of caffeine can therefore be used in healthy volunteers to mimic the effects of insomnia and thus to test the hypnotic effects of medication. This study assessed the effects of caffeine on sleep architecture and electroencephalography (EEG) spectrum alone and in combination with two different sleep-promoting medications. Home polysomnography was performed in 12 healthy male volunteers in a double-blind study whereby subjects received placebo, caffeine (150 mg), caffeine plus zolpidem (10 mg) and caffeine plus trazodone (100 mg) at bedtime in a randomised crossover design. In addition to delaying sleep onset, caffeine decreased total sleep time (TST), sleep efficiency (SE) and stage 2 sleep without significantly altering wake after sleep onset or the number of awakenings. Zolpidem attenuated the caffeine-induced decrease in SE and increased spindle density in the caffeine plus zolpidem combination compared with placebo. Trazodone attenuated the decrease in SE and TST, and it also increased stage 3 sleep, decreased the number of awakenings and decreased the spindle density. No significant changes in rapid eye movement (REM) sleep were observed, neither was any significant alteration in slow wave activity nor other EEG spectral measures, although the direction of change was similar to that previously reported for caffeine and appeared to 'normalise' after trazodone. These data suggest that caffeine mimics some, but not all of the sleep disruption seen in insomnia and that its disruptive effects are differentially attenuated by the actions of sleep-promoting compounds with distinct mechanisms of action.


Subject(s)
Caffeine/toxicity , Central Nervous System Stimulants/toxicity , Hypnotics and Sedatives/pharmacology , Sleep Stages/drug effects , Adult , Cross-Over Studies , Double-Blind Method , Electroencephalography , Humans , Male , Polysomnography , Pyridines/pharmacology , Sleep Initiation and Maintenance Disorders/chemically induced , Sleep Initiation and Maintenance Disorders/drug therapy , Trazodone/pharmacology , Young Adult , Zolpidem
6.
Psychopharmacology (Berl) ; 204(3): 499-509, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19205667

ABSTRACT

BACKGROUND: The selective mGluR5 antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP) is reported to be anxiolytic in several animal models of anxiety, including the conditioned emotional response (CER) paradigm. Suppression of responding during conditioned stimulus (CS) presentation in CER may reflect behavioural competition between lever pressing and adopting a shock-avoidance posture, or it may alternatively reflect altered value of the food reward following its association with a footshock, thus reducing its ability to motivate responding. If this is the case, then drugs that reduce the CER may interfere with the mechanism by which CSs are able to motivate responding, rather than by reducing anxiety. The standard test of the ability of Pavlovian cues to motivate responding is the Pavlovian-to-instrumental transfer (PIT) paradigm and it has recently been suggested that CER may be 'negative PIT'. MATERIALS AND METHODS: We compared the effect of MPEP (0, 3, 10 and 30 mg/kg) and diazepam (0, 1, 3 and 10 mg/kg) in CER and PIT. RESULTS: Both MPEP and diazepam significantly reduced conditioned suppression in the CER paradigm. MPEP, but not diazepam, significantly reduced PIT. CONCLUSION: The findings support the hypothesis that MPEP may reduce expression of anxiety in the CER paradigm by interfering with the way in which emotionally salient cues are able to affect behaviour, but do not support such an analysis of the effect of diazepam. Diazepam and MPEP may therefore achieve their effects in CER by influencing different psychological processes.


Subject(s)
Anti-Anxiety Agents/pharmacology , Conditioning, Operant/drug effects , Diazepam/pharmacology , Emotions/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Pyridines/pharmacology , Receptors, Kainic Acid/antagonists & inhibitors , Animals , Cues , Dose-Response Relationship, Drug , Food , Habituation, Psychophysiologic/drug effects , Male , Motivation , Rats
7.
Schizophr Res ; 102(1-3): 283-94, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18400471

ABSTRACT

The NMDA receptor co-agonists D-serine and glycine are thought to contribute to glutamatergic dysfunction in schizophrenia. They are removed from the synapse by specific neuronal and glial transporters, the status of which is clearly relevant to theories of D-serine and glycine function in the disorder. D-serine is primarily transported by Asc-1, and glycine by GlyT1 but maybe also by SNAT2. As a first step to addressing this issue, we studied Asc-1, GlyT1 and SNAT2 expression in dorsolateral prefrontal cortex and cerebellum of 18 subjects with schizophrenia and 20 controls, using immunoblotting and in situ hybridization. Asc-1 protein and SNAT2 mRNA were decreased in schizophrenia in both regions. GlyT1 mRNA and protein, and Asc-1 mRNA, were not altered. Antipsychotic administration for 14 days did not alter expression of the genes in rat brain. Unchanged GlyT1 suggests that glycine transport is not markedly affected in schizophrenia, and therefore that increased synaptic removal is not the basis for the putative deficit in glycine modulation of NMDA receptors in the disorder. Lowered Asc-1 in schizophrenia implies that D-serine reuptake is reduced, perhaps as a response to decreased synaptic D-serine availability. However, this interpretation remains speculative. Further investigations will be valuable in the evaluation of these transporters as potential therapeutic targets in psychosis.


Subject(s)
Cerebellum/metabolism , Glutamates/physiology , Glycine Plasma Membrane Transport Proteins/metabolism , Glycine/metabolism , Prefrontal Cortex/metabolism , Schizophrenia/physiopathology , Serine/metabolism , Amino Acid Transport System A/genetics , Amino Acid Transport System A/metabolism , Animals , Antipsychotic Agents/pharmacology , Blotting, Western , Control Groups , Female , Gene Expression/drug effects , Glutamates/genetics , Glutamates/metabolism , Glycine/genetics , Glycine Plasma Membrane Transport Proteins/genetics , Humans , In Situ Hybridization , Male , Middle Aged , Neurons/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/genetics , Schizophrenia/metabolism , Serine/genetics , Synapses/drug effects , Synapses/metabolism
8.
Psychopharmacology (Berl) ; 197(4): 591-600, 2008 May.
Article in English | MEDLINE | ID: mdl-18264695

ABSTRACT

RATIONALE: Tiagabine is an anticonvulsant drug which may also have sleep-enhancing properties. It acts by inhibiting reuptake at the gamma-aminobutyric acid (GABA) transporter (GAT-1). OBJECTIVES: The aim of the study was to determine whether tiagabine acted as a discriminative stimulus and, if so, whether other GABAergic compounds would generalise to it. MATERIALS AND METHODS: Rats were trained to discriminate tiagabine (30 mg/kg p.o.) from vehicle, and generalisation to drugs that modulate GABA was assessed. RESULTS: Gaboxadol (5-20 mg/kg p.o.), a selective extrasynaptic GABA A agonist, generalised to tiagabine, although the extent of the generalisation was inconclusive. Indiplon (1 mg/kg p.o.), a benzodiazepine-like hypnotic, also partially generalised to tiagabine, although zolpidem and S-zopiclone did not. Baclofen, a GABA B receptor agonist, and gabapentin, which increases synaptic GABA, did not generalise to tiagabine. (+)-Bicuculline (3 mg/kg i.p.), a GABA A receptor antagonist, blocked the tiagabine cue, but the less brain-penetrant salt form, bicuculline methochloride, had no effect. CONCLUSIONS: These data suggest that tiagabine generates a discriminative stimulus in rats, and provides a central GABA-mediated cue, but is distinct from the other GABAergic compounds tested.


Subject(s)
Anticonvulsants/pharmacology , Discrimination Learning/drug effects , GABA Agents/pharmacology , Nipecotic Acids/pharmacology , Administration, Oral , Amines/pharmacology , Animals , Azabicyclo Compounds/pharmacology , Baclofen/pharmacology , Benzodiazepines/pharmacology , Bicuculline/analogs & derivatives , Bicuculline/pharmacology , Brain/drug effects , Cyclohexanecarboxylic Acids/pharmacology , Dose-Response Relationship, Drug , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , GABA Plasma Membrane Transport Proteins/drug effects , Gabapentin , Generalization, Stimulus/drug effects , Hypnotics and Sedatives/pharmacology , Injections, Intraperitoneal , Isoxazoles/pharmacology , Male , Piperazines/pharmacology , Pyridines/pharmacology , Rats , Thiophenes/pharmacology , Tiagabine , Zolpidem , gamma-Aminobutyric Acid/pharmacology
9.
Mol Psychiatry ; 13(5): 498-506, 2008 May.
Article in English | MEDLINE | ID: mdl-17622221

ABSTRACT

Considerable attention has focused on regulation of central tryptophan hydroxylase (TPH) activity and protein expression. At the time of these earlier studies, it was thought that there was a single central TPH isoform. However, with the recent identification of TPH2, it becomes important to distinguish between regulatory effects on the protein expression and activity of the two isoforms. We have generated a TPH2-specific polyclonal antiserum (TPH2-6361) to study regulation of TPH2 at the protein level and to examine the distribution of TPH2 expression in rodent and human brain. TPH2 immunoreactivity (IR) was detected throughout the raphe nuclei, in lateral hypothalamic nuclei and in the pineal body of rodent and human brain. In addition, a prominent TPH2-IR fiber network was found in the human median eminence. We recently reported that glucocorticoid treatment of C57/Bl6 mice for 4 days markedly decreased TPH2 messenger RNA levels in the raphe nuclei, whereas TPH1 mRNA was unaffected. The glucocorticoid-elicited inhibition of TPH2 gene expression was blocked by co-administration of the glucocorticoid receptor antagonist mifepristone (RU-486). Using TPH2-6361, we have extended these findings to show a dose-dependent decrease in raphe TPH2 protein levels in response to 4 days of treatment with dexamethasone; this effect was blocked by co-administration of mifepristone. Moreover, the glucocorticoid-elicited inhibition of TPH2 was functionally significant: serotonin synthesis was significantly reduced in the frontal cortex of glucocorticoid-treated mice, an effect that was blocked by mifepristone co-administration. This study provides further evidence for the glucocorticoid regulation of serotonin biosynthesis via inhibition of TPH2 expression, and suggest that elevated glucocorticoid levels may be relevant to the etiology of psychiatric diseases, such as depression, where hypothalamic-pituitary-adrenal axis dysregulation has been documented.


Subject(s)
5-Hydroxytryptophan/biosynthesis , Dexamethasone/analogs & derivatives , Frontal Lobe/chemistry , Nerve Tissue Proteins/biosynthesis , Raphe Nuclei/enzymology , Tryptophan Hydroxylase/analysis , Tryptophan Hydroxylase/biosynthesis , 5-Hydroxytryptophan/analysis , Amino Acid Sequence , Animals , Antibody Specificity , Dexamethasone/pharmacology , Enzyme Induction/drug effects , Female , Frontal Lobe/drug effects , Humans , Immune Sera , Mice , Mice, Inbred C57BL , Mifepristone/pharmacology , Molecular Sequence Data , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Ovariectomy , Peptide Fragments/immunology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/immunology , RNA, Messenger/biosynthesis , Raphe Nuclei/drug effects , Rats , Rats, Sprague-Dawley , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/immunology
10.
Neuropharmacology ; 52(3): 844-53, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17196996

ABSTRACT

Gaboxadol is a selective extrasynaptic GABA(A) receptor agonist (SEGA) which enhances slow-wave sleep, and may act principally at extrasynaptic GABA(A)alpha4betadelta receptors. Drug discrimination is a very useful approach for exploring in vivo pharmacological similarities and differences between compounds and was therefore used to compare gaboxadol and zolpidem, an established hypnotic drug, against zopiclone, S-zopiclone, indiplon and tiagabine, all of which have been reported to enhance sleep. Gaboxadol generalised to itself, but not to zolpidem, zopiclone, S-zopiclone, R-zopiclone, indiplon or tiagabine. By contrast, zolpidem generalised to itself, zopiclone, S-zopiclone and indiplon, but not to R-zopiclone (the inactive enantiomer of zopiclone), gaboxadol or tiagabine. This suggests that zolpidem, zopiclone, S-zopiclone and indiplon share a discriminative stimulus, which may be mediated by their efficacy at GABA(A)alpha1betagamma receptors. Gaboxadol and tiagabine each have a different discriminative stimulus from all the other drugs tested.


Subject(s)
Discrimination Learning/drug effects , GABA Agonists/pharmacology , Generalization, Psychological/drug effects , Isoxazoles/pharmacology , Sleep/drug effects , Animals , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Drug Evaluation , Drug Interactions , Male , Rats , Rats, Sprague-Dawley
11.
J Psychopharmacol ; 21(6): 597-602, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17092981

ABSTRACT

Beta3 adrenoceptor agonists show an antidepressant-like profile in preclinical rodent assays and improve mood in clinically-obese patients. These observations suggest a possible antidepressant utility for beta3 adrenoceptor agonists. The present study examined the effects of acute and chronic administration of the beta3 adrenoceptor agonist CL 316243 on two physiological indicators of antidepressant activity in the rat: hypothalamic 5-HT synthesis and suppression of REM sleep. 5-HT synthesis was estimated by the accumulation of 5-hydroxytryptophan (5-HTP) after treatment with the L-aromatic acid decarboxylase inhibitor NSD 1015. Sleep-wake patterns were monitored using electroencephalogram and electromyogram signals collected by radiotelemetry. Rats were administered CL 316243 acutely or once daily for 11 days. Acute administration of CL 316243 significantly increased hypothalamic 5-HT synthesis, as indicated by increased levels of 5-HTP, and reduced the amount of REM sleep. However, chronic administration produced no changes in 5-HTP or REM compared with vehicle treatment. The present observations suggest that acute administration of CL 316243 causes antidepressant-like effects on REM sleep, possibly mediated by increased central 5-HT synthesis. However, these effects are not maintained with repeated dosing.


Subject(s)
Adrenergic beta-3 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Antidepressive Agents/pharmacology , Dioxoles/pharmacology , Hypothalamus/drug effects , Serotonin/biosynthesis , Sleep, REM/drug effects , 5-Hydroxytryptophan/biosynthesis , Animals , Aromatic Amino Acid Decarboxylase Inhibitors , Aromatic-L-Amino-Acid Decarboxylases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Hydrazines/pharmacology , Hypothalamus/enzymology , Hypothalamus/metabolism , Male , Rats , Receptors, Adrenergic, beta-3/metabolism , Time Factors , Wakefulness/drug effects
12.
Mol Cell Neurosci ; 32(4): 324-34, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16843004

ABSTRACT

D-amino acid oxidase (DAO) degrades D-serine, a co-agonist at the NMDA receptor (NMDAR). Hypofunction of the NMDAR has been suggested to contribute to the pathophysiology of schizophrenia. Intriguingly, DAO has been recently identified as a risk factor for schizophrenia through genetic association studies. A naturally occurring mouse strain (ddY/DAO-) has been identified which lacks DAO activity. We have characterized this strain both behaviorally and biochemically to evaluate DAO as a target for schizophrenia. We have confirmed that this strain lacks DAO activity and shown for the first time it has increased occupancy of the NMDAR glycine site due to elevated extracellular D-serine levels and has enhanced NMDAR function in vivo. Furthermore, the ddY/DAO- strain displays behaviors which suggest that it will be a useful tool for evaluation of the clinical benefit of DAO inhibition in schizophrenia.


Subject(s)
Brain Chemistry/genetics , D-Amino-Acid Oxidase/deficiency , Mice, Knockout/physiology , Schizophrenia/physiopathology , Acoustic Stimulation/methods , Animals , Brain Chemistry/drug effects , Cyclic GMP/metabolism , Disease Models, Animal , Dose-Response Relationship, Radiation , Excitatory Amino Acid Antagonists/pharmacology , Extremities/physiology , Female , Male , Mice , Mice, Inbred Strains , Motor Activity/drug effects , Motor Activity/genetics , Neural Inhibition/genetics , Neurologic Examination/methods , Phencyclidine/administration & dosage , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Quinolones/pharmacology , Reaction Time/genetics , Reflex, Startle/genetics , Schizophrenia/metabolism , Sex Factors , Swimming/physiology
13.
Neuropharmacology ; 48(7): 1002-11, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15857627

ABSTRACT

(2S)-2-{[(3,5-Diflurophenyl)acetyl]amino}-N-[(3S)-1-methyl-2-oxo-5-phenyl-2,3-dihydro-1H-1,4-benzodiazepin-3-yl]propanamide (compound E) is a gamma-secretase inhibitor capable of reducing amyloid beta-peptide (1-40) and amyloid beta-peptide (1-42) levels. In this study we investigated the effect of in vivo administration of compound E on guinea-pig plasma, CSF and cortical amyloid beta-peptide (1-40) concentration. Using repeated sampling of CSF, compound E (30 mg/kg p.o.) was shown to cause a time-dependent decrease in CSF amyloid beta-peptide (1-40) levels, which was maximal at 3 h (70% inhibition), compared to baseline controls. After 3 h administration, compound E (3, 10 and 30 mg/kg p.o.), reduced plasma, CSF and DEA-extracted cortical amyloid beta-peptide (1-40) levels by 95, 97 and 99%; 26, 48 and 78%; 32, 33, and 47%, respectively, compared to vehicle control values. In the same animals, compound E (3, 10 and 30 mg/kg p.o.) inhibited cortical gamma-secretase activity, determined ex vivo using the recombinant substrate C100Flag, by 40, 71 and 79% of controls, respectively. These data demonstrate the value of determining not only the extent by which systemic administration of a gamma-secretase inhibitor reduces amyloid beta-peptide, but also the inhibition of brain gamma-secretase activity, as a more direct estimate of enzyme occupancy.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Protease Inhibitors/administration & dosage , Administration, Oral , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/metabolism , Animals , Arsenicals/administration & dosage , Bisbenzimidazole/administration & dosage , Bisbenzimidazole/analogs & derivatives , Dose-Response Relationship, Drug , Endopeptidases , Enzyme Activation/drug effects , Enzyme Activation/physiology , Guinea Pigs , Male , Peptide Fragments/metabolism
14.
Neuropharmacology ; 48(4): 492-502, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15755477

ABSTRACT

The affinity of several antidepressant and antipsychotic drugs for the 5-HT7 receptor and its CNS distribution suggest potential in the treatment of psychiatric diseases. However, there is little direct evidence of receptor function in vivo to support this. We therefore evaluated 5-HT7 receptors as a potential drug target by generating and assessing a 5-HT7 receptor knockout mouse. No difference in assays sensitive to potential psychotic or anxiety states was observed between the 5-HT7 receptor knockout mice and wild type controls. However, in the Porsolt swim test, 5-HT7 receptor knockout mice showed a significant decrease in immobility compared to controls, a phenotype similar to antidepressant treated mice. Intriguingly, treatment of wild types with SB-258719, a selective 5-HT7 receptor antagonist, did not produce a significant decrease in immobility unless animals were tested in the dark (or active) cycle, rather than the light, adding to the body of evidence suggesting a circadian influence on receptor function. Extracellular recordings from hypothalamic slices showed that circadian rhythm phase shifts to 8-OH-DPAT are attenuated in the 5-HT7 receptor KO mice also indicating a role for the receptor in the regulation of circadian rhythms. These pharmacological and genetic knockout studies provide the first direct evidence that 5-HT7 receptor antagonists should be investigated for efficacy in the treatment of depression.


Subject(s)
Depressive Disorder/drug therapy , Depressive Disorder/genetics , Receptors, Serotonin/genetics , Serotonin Antagonists/therapeutic use , Animals , Gene Targeting/methods , Immobilization/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Piperidines/pharmacology , Piperidines/therapeutic use , Receptors, Serotonin/deficiency , Reflex, Startle/drug effects , Reflex, Startle/physiology , Serotonin Antagonists/pharmacology , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
15.
Neuropharmacology ; 44(8): 1031-7, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12763096

ABSTRACT

The 5-HT(7) receptor is a recent addition to the 5-HT receptor family and to date there is no clear idea as to its potential role in the CNS. The receptor has been mapped by in situ hybridization and 5-HT(7)-like immunoreactivity and has been detected in discrete areas of the brain including the hypothalamus (Oliver et al., 1999). This suggests the receptor may be involved in temperature regulation and have shown that a selective 5-HT(7) receptor antagonist reverses the hypothermic effect of 5-CT in guinea-pigs. The current study confirmed that the 5-HT(7) receptor antagonists, SB-269970 (1-30 mg/kg, i.p.) and SB-258719 (5-20 mg/kg, i.p.), but not the 5-HT(1A) receptor antagonist, WAY 100635(0.1-1 mg/kg, s.c.), or the 5-HT(1B/D) antagonist, GR127935 (1.25-5 mg/kg, i.p.), reversed the hypothermic effect of 5-CT in mice. In addition the effect of 5-CT on body temperature was examined on 5-HT(7) receptor null mutant mice. 5-CT (0.1-1 mg/kg, i.p.) significantly reduced rectal temperature in wildtype but not 5-HT(7) receptor knockout mice. This suggests that the hypothermic effects of 5-CT are mediated through the 5-HT(7) receptor. All procedures were carried out in accordance with the UK Animals (Scientific Procedures) Act (1986).


Subject(s)
Hypothermia/metabolism , Receptors, Serotonin/drug effects , Serotonin Receptor Agonists/pharmacology , Serotonin/analogs & derivatives , Serotonin/pharmacology , Animals , Body Temperature/drug effects , Hypothermia/chemically induced , Hypothermia/physiopathology , Injections, Intraventricular , Mice , Mice, Knockout , Phenols/pharmacology , Piperidines/pharmacology , Receptors, Serotonin/genetics , Serotonin Antagonists/pharmacology , Sulfonamides/pharmacology
16.
Neuropharmacology ; 44(4): 439-48, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12646281

ABSTRACT

This study examined the long-term change in serotonergic (5-hydroxytryptamine, 5-HT) neuronal function and 5-HT(2C) receptor agonist-induced behaviour following treatment of young rats with 3,4-methylenedioxymethamphetamine (MDMA). On post-natal day (PND) 28, Lister-hooded rats received either MDMA (15 mg/kg i.p.) or saline (1 ml/kg i.p.) twice daily for 3 days. On PND 50 social interaction was assessed between treatment-matched pairs of rats derived from separate litters. The effect of either the 5-HT(2C) receptor agonist, m-chlorophenylpiperazine (m-CPP, 2.5 or 1 mg/kg i.p., respectively) or saline was examined on open-field exploration (PND 52) and elevated plus-maze behaviour (PND 56). Acutely, MDMA produced hyperlocomotion and hypothermia compared with saline injection (p<0.001). Following 20 days abstinence, social interaction was decreased by 26% (p<0.05) in MDMA pre-treated rats compared with saline controls, without any change in locomotion. There was no difference in open-field or elevated plus-maze behaviour between pre-treatment groups. m-CPP caused hypolocomotion in the open-field and decreased both the percentage entries into, and time spent in, the open arms of the elevated plus-maze to a comparable extent in MDMA and saline pre-treated rats. Hippocampal and frontal cortical 5-HT and 5-hydroxyindoleacetic acid (5-HIAA) were significantly reduced in MDMA pre-treated rats, without any change in [(3)H]paroxetine binding or plasma corticosterone levels. These data suggest that the MDMA-induced reduction in social interaction is not mediated via alteration of 5-HT(2C) receptor function.


Subject(s)
N-Methyl-3,4-methylenedioxyamphetamine/adverse effects , Receptors, Serotonin/drug effects , Serotonin Agents/adverse effects , Social Behavior , Animals , Brain/drug effects , Brain/metabolism , Corticosterone/blood , Exploratory Behavior/drug effects , Hydroxyindoleacetic Acid/metabolism , Hypothermia/chemically induced , In Vitro Techniques , Male , Maze Learning/drug effects , Motor Activity/drug effects , Paroxetine/metabolism , Piperazines/pharmacology , Radioligand Assay , Rats , Receptor, Serotonin, 5-HT2C , Serotonin/metabolism , Serotonin Receptor Agonists/pharmacology , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology
17.
Brain Res Brain Res Protoc ; 8(3): 191-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11733195

ABSTRACT

An alpha(1) adrenoceptor (alpha(1)-AdR) assay using [(3)H]prazosin binding in mouse brain is described which allows in vivo determination of central alpha(1)-AdR occupancy for ligands with alpha(1)-AdR affinity. Binding of [3H]prazosin in rat and mouse brain membranes in vitro was used to characterise the pharmacological profile of alpha(1)-AdRs in order to determine any potential species variations. Saturation and displacement studies yielded comparable affinity and pharmacological profile for [(3)H]prazosin binding in mouse and rat brain homogenates. These studies confirmed the absence of species variation for ligands in central alpha(1)-AdR pharmacology which is in good agreement with previous studies in rat brain. Subsequently, in vivo binding of [(3)H]prazosin in mouse whole brain was used to measure the occupancy of a number of AdR ligands. Timecourse studies revealed that a [3H]prazosin (5 mu Ci/mouse) pretreatment time of at least 20 min following intravenous (i.v.) administration was required for optimal specific binding. Ligands were administered systemically 40 min prior to i.v. administration of radiolabel. The alpha(1)-adrenoceptor ligands prazosin (ED(50)=0.15 mg/kg i.p.), benoxathian (0.52 mg/kg i.p.) and phentolamine (51 mg/kg i.p.) were all able to block in vivo [(3)H]prazosin binding from mouse brain. In addition, receptor occupancy values for a number of compounds including haloperidol (ED(50)=0.83 mg/kg s.c.), clozapine (2.2 mg/kg s.c.) and MDL-100907 [R(+)-alpha-(2,3-dimethoxyphenyl)-1-[2-(4-fluorophenylethyl)]-4-piperidine-methanol], (10 mg/kg s.c.)], which possess high to moderate affinity at alpha(1)-adrenoceptors, were also determined. These results suggest that in the mouse, [(3)H]prazosin binding can be used to measure in vivo receptor occupancy of ligands with affinity at central alpha(1)-adrenoceptors.


Subject(s)
Adrenergic alpha-Antagonists , Central Nervous System/metabolism , Prazosin , Receptors, Adrenergic, alpha-1/metabolism , Animals , Binding, Competitive/drug effects , Dose-Response Relationship, Drug , Male , Membranes/metabolism , Mice , Mice, Inbred Strains , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/drug effects , Species Specificity
18.
J Med Chem ; 44(10): 1603-14, 2001 May 10.
Article in English | MEDLINE | ID: mdl-11334570

ABSTRACT

The development of very high affinity, selective, and bioavailable h5-HT(2A) receptor antagonists is described. By investigation of the optimal position for the basic nitrogen in a series of 2-phenyl-3-piperidylindoles, it was found that with the basic nitrogen at the 3-position of the piperidine it was not necessary to further substitute the piperidine in order to obtain good binding at h5-HT(2A) receptors. This meant the compounds no longer had high affinity at the IKr potassium channel, an issue with previous series of 2-aryl-3-(4-piperidyl)indoles. Improvements could be made to oral bioavailability in this series by reduction of the pK(a) of the basic nitrogen, by adding a fluorine atom to the piperidine ring, leading to 3-(4-fluoropiperidin-3-yl)-2-phenyl-1H-indole (17). Metabolic studies with this compound identified oxidation at the 6-position of the indole as a major route in vitro and in vivo in rats. Blocking this position with a fluorine atom led to 6-fluoro-3-(4-fluoropiperidin-3-yl)-2-phenyl-1H-indole (22), an antagonist with 0.06 nM affinity for h5-HT(2A) receptors, with bioavailability of 80% and half-life of 12 h in rats.


Subject(s)
Indoles/chemical synthesis , Piperidines/chemical synthesis , Receptors, Serotonin/drug effects , Serotonin Antagonists/chemical synthesis , Animals , Binding, Competitive , Biological Availability , CHO Cells , Cerebral Cortex/metabolism , Cricetinae , Dogs , Female , Humans , In Vitro Techniques , Indoles/chemistry , Indoles/metabolism , Indoles/pharmacology , Male , Microsomes, Liver/metabolism , Oxidation-Reduction , Piperidines/chemistry , Piperidines/metabolism , Piperidines/pharmacology , Potassium Channels/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A , Receptors, Serotonin/metabolism , Serotonin Antagonists/chemistry , Serotonin Antagonists/metabolism , Serotonin Antagonists/pharmacology , Structure-Activity Relationship
20.
J Neurochem ; 75(6): 2455-63, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11080197

ABSTRACT

We have performed [(3)H]ifenprodil binding experiments under NMDA receptor-specific assay conditions to provide the first detailed characterisation of the pharmacology of the ifenprodil site on NMDA NR1/NR2B receptors, using recombinant human NR1a/NR2B receptors stably expressed in L(tk-) cells, in comparison with rat cortex/hippocampus membranes. [(3)H]Ifenprodil bound to a single, saturable site on both human recombinant NR1a/NR2B receptors and native rat receptors with B:(max) values of 1.83 and 2.45 pmol/mg of protein, respectively, and K:(D) values of 33.5 and 24.8 nM:, respectively. The affinity of various ifenprodil site ligands-eliprodil, (R:(*), R:(*))-4-hydroxy-alpha-(4-hydroxyphenyl)-beta-methyl-4-pehnyl-1-pi per idineethanol [(+/-)-CP-101,606], cis-3-[4-(4-fluorophenyl)-4-hydroxy-1-piperidinyl]-3, 4-dihydro-2H:-1-benzopyran-4,7-diol [(+/-)-CP-283,097], and (R:(*), S:(*))-alpha-(4-hydroxyphenyl)-beta-methyl-4-(phenylmethyl)-1-piperid inepropanol [(+/-)-Ro 25-6981] was very similar for inhibition of [(3)H]ifenprodil binding to recombinant human NR1a/NR2B and native rat receptors, whereas allosteric inhibition of [(3)H]ifenprodil binding by polyamine site ligands (spermine, spermidine, and arcaine) showed approximately twofold lower affinity for recombinant receptors compared with native receptors. Glutamate site ligands were less effective at modulating [(3)H]ifenprodil binding to recombinant NR1a/NR2B receptors compared with native rat receptors. The NMDA receptor-specific [(3)H]ifenprodil binding conditions described were also applied to ex vivo experiments to determine the receptor occupancy of ifenprodil site ligands [ifenprodil, (+/-)-CP-101,606, (+/-)-CP-283,097, and (+/-)-Ro 25-6981] given systemically.


Subject(s)
Cell Membrane/metabolism , Piperidines/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Allosteric Site/drug effects , Animals , Binding Sites/drug effects , Binding, Competitive/drug effects , Brain Chemistry , Cell Line , Dopamine Uptake Inhibitors/pharmacology , Dose-Response Relationship, Drug , Humans , Male , Mice , Prosencephalon/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...