Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters











Publication year range
1.
Article in English | MEDLINE | ID: mdl-34927075

ABSTRACT

Orexin receptors (nomenclature as agreed by the NC-IUPHAR Subcommittee on Orexin receptors [42]) are activated by the endogenous polypeptides orexin-A and orexin-B (also known as hypocretin-1 and -2; 33 and 28 aa) derived from a common precursor, preproorexin or orexin precursor, by proteolytic cleavage and some typical peptide modifications [109]. Currently the only orexin receptor ligands in clinical use are suvorexant and lemborexant, which are used as hypnotics. Orexin receptor crystal structures have been solved [134, 133, 54, 117, 46].

2.
Nat Rev Drug Discov ; 11(11): 833-46, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23123941

ABSTRACT

Existing drugs for Alzheimer's disease provide symptomatic benefit for up to 12 months, but there are no approved disease-modifying therapies. Given the recent failures of various novel disease-modifying therapies in clinical trials, a complementary strategy based on repositioning drugs that are approved for other indications could be attractive. Indeed, a substantial body of preclinical work indicates that several classes of such drugs have potentially beneficial effects on Alzheimer's-like brain pathology, and for some drugs the evidence is also supported by epidemiological data or preliminary clinical trials. Here, we present a formal consensus evaluation of these opportunities, based on a systematic review of published literature. We highlight several compounds for which sufficient evidence is available to encourage further investigation to clarify an optimal dose and consider progression to clinical trials in patients with Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Drug Approval , Drug Design , Alzheimer Disease/physiopathology , Animals , Clinical Trials as Topic , Drug Evaluation, Preclinical , Drug Industry/economics , Humans
3.
Behav Neurosci ; 126(3): 488-92, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22642890

ABSTRACT

Prader-Willi syndrome (PWS) is caused by lack of paternally derived gene expression from the imprinted gene cluster on human chromosome 15q11-q13. PWS is characterized by severe hypotonia, a failure to thrive in infancy and, on emerging from infancy, evidence of learning disabilities and overeating behavior due to an abnormal satiety response and increased motivation by food. We have previously shown that an imprinting center deletion mouse model (PWS-IC) is quicker to acquire a preference for, and consume more of a palatable food. Here we examined how the use of this palatable food as a reinforcer influences learning in PWS-IC mice performing a simple appetitive learning task. On a nonspatial maze-based task, PWS-IC mice acquired criteria much quicker, making fewer errors during initial acquisition and also reversal learning. A manipulation where the reinforcer was devalued impaired wild-type performance but had no effect on PWS-IC mice. This suggests that increased motivation for the reinforcer in PWS-IC mice may underlie their enhanced learning. This supports previous findings in PWS patients and is the first behavioral study of an animal model of PWS in which the motivation of behavior by food rewards has been examined.


Subject(s)
Appetitive Behavior/physiology , Motivation/genetics , Prader-Willi Syndrome/genetics , Reversal Learning/physiology , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prader-Willi Syndrome/psychology
4.
J Neurosci Res ; 89(8): 1218-27, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21538462

ABSTRACT

The molecular basis of schizophrenia is poorly understood; however, different brain regions are believed to play distinct roles in disease symptomology. We have studied gene expression in the superior temporal cortex (Brodmann area 22; BA22), which may play a role in positive pathophysiology, and compared our results with data from the anterior prefrontal cortex (BA10), which shows evidence for a role in negative symptoms. Genome-wide mRNA expression was determined in the BA22 region in 23 schizophrenics and 19 controls and compared with a BA10 data set from the same subjects. After adjustments for confounding sources of variation, we carried out GeneGO pathway enrichment analysis in each region. Significant differences were seen in age-related transcriptional changes between the BA22 and the BA10 regions, 21.8% and 41.4% of disease-associated transcripts showing age association, respectively. After removing age associated changes from our data, we saw the highest enrichment in processes mediating cell adhesion, synaptic contact, cytoskeletal remodelling, and apoptosis in the BA22 region. For the BA10 region, we observed the strongest changes in reproductive signalling, tissue remodelling, and cell differentiation. Further exploratory analysis also identified potentially disease-relevant processes that were undetected in our more stringent primary analysis, including autophagy in the BA22 region and the amyloid process in the BA10 region. Collectively, our analysis suggests disruption of many common pathways and processes underpinning synaptic plasticity in both regions in schizophrenia, whereas individual regions emphasize changes in certain pathways that may help to highlight pathway-specific therapeutic opportunities to treat negative or positive symptoms of the disease.


Subject(s)
Prefrontal Cortex/metabolism , Schizophrenia/genetics , Temporal Lobe/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Female , Gene Expression , Gene Expression Profiling , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Schizophrenia/metabolism
5.
Neuropharmacology ; 58(8): 1206-14, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20206188

ABSTRACT

N-desmethylclozapine (NDMC) has been reported to display partial agonism at the human recombinant and rat native M(1) mAChR, a property suggested to contribute to the clinical efficacy of clozapine. However, the profile of action of NDMC at the human native M(1) mAChR has not been reported. The effect of NDMC on M(1) mAChR function was investigated in human native tissues by assessing its effect on (1) M(1) mAChR-mediated stimulation of [(35)S]-GTPgammaS-G(q/11)alpha binding to human post mortem cortical membranes and (2) the M(1) mAChR-mediated increase in neuronal firing in human neocortical slices. NDMC displayed intrinsic activities of 46+/-9%, compared to oxo-M, at the human recombinant M(1) receptor, in FLIPR studies and 35+/-4% at rat native M(1) receptors in [(35)S]-GTPgammaS-G(q/11)alpha binding studies. In [(35)S]-GTPgammaS-G(q/11)alpha binding studies in human cortex, oxo-M stimulated binding by 240+/-26% above basal with a pEC(50) of 6.56+/-0.05. In contrast, NDMC did not stimulate [(35)S]-GTPgammaS-G(q/11)alpha binding to human cortical membranes but antagonised the response to oxo-M (2microM) showing a pK(B) of 6.8, comparable to its human recombinant M(1) mAChR affinity (pK(i)=6.9) derived from [(3)H]-NMS binding studies. In human, contrary to the rat neocortical slices, NDMC did not elicit a significant increase in M(1) mAChR-mediated neuronal firing, and attenuated a carbachol-induced increase in neuronal firing when pre-applied. These data indicate that, whereas NDMC displays moderate to low levels of partial agonism at the human recombinant and rat native M(1) mAChR, respectively, it acts as an antagonist at the M(1) mAChR in human cortex.


Subject(s)
Clozapine/analogs & derivatives , Receptor, Muscarinic M1/antagonists & inhibitors , Action Potentials , Animals , Calcium/metabolism , Clozapine/pharmacology , Drug Partial Agonism , Hippocampus/drug effects , Hippocampus/physiology , Humans , In Vitro Techniques , Neocortex/drug effects , Neocortex/physiology , Oxotremorine/analogs & derivatives , Oxotremorine/pharmacology , Protein Binding , Radioligand Assay , Rats , Receptor, Muscarinic M1/agonists , Recombinant Proteins/agonists
6.
Eur J Neurosci ; 31(1): 156-64, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20092561

ABSTRACT

The genes in the imprinted cluster on human chromosome 15q11-q13 are known to contribute to psychiatric conditions such as schizophrenia and autism. Major disruptions of this interval leading to a lack of paternal allele expression give rise to Prader-Willi syndrome (PWS), a neurodevelopmental disorder with core symptoms of a failure to thrive in infancy and, on emergence from infancy, learning disabilities and over-eating. Individuals with PWS also display a number of behavioural problems and an increased incidence of neuropsychiatric abnormalities, which recent work indicates involve aspects of frontal dysfunction. To begin to examine the contribution of genes in this interval to relevant psychological and behavioural phenotypes, we exploited the imprinting centre (IC) deletion mouse model for PWS (PWS-IC(+/-)) and the five-choice serial reaction time task (5-CSRTT), which is primarily an assay of visuospatial attention and response control that is highly sensitive to frontal manipulations. Locomotor activity, open-field behaviour and sensorimotor gating were also assessed. PWS-IC(+/-) mice displayed reduced locomotor activity, increased acoustic startle responses and decreased prepulse inhibition of startle responses. In the 5-CSRTT, the PWS-IC(+/-) mice showed deficits in discriminative response accuracy, increased correct reaction times and increased omissions. Task manipulations confirmed that these differences were likely to be due to impaired attention. Our data recapitulate several aspects of the PWS clinical condition, including findings consistent with frontal abnormalities, and may indicate novel contributions of the imprinted genes found in 15q11-q13 to behavioural and cognitive function generally.


Subject(s)
Cognition Disorders/genetics , Exploratory Behavior , Motor Activity/genetics , Prader-Willi Syndrome/genetics , Animals , Attention , Body Weight , Brain/physiopathology , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Discrimination, Psychological , Disease Models, Animal , Exploratory Behavior/physiology , Failure to Thrive/genetics , Failure to Thrive/physiopathology , Female , Genomic Imprinting , Male , Mice , Mice, Transgenic , Motor Activity/physiology , Neuropsychological Tests , Prader-Willi Syndrome/physiopathology , Prader-Willi Syndrome/psychology , Reaction Time , Reflex, Startle/genetics , Reflex, Startle/physiology , Sequence Deletion
7.
Eur J Pharmacol ; 627(1-3): 106-14, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-19879867

ABSTRACT

Clinical evaluation of tachykinin NK(3) receptor antagonists has provided support for the therapeutic utility of this target in schizophrenia. However, these studies have not been entirely conclusive, possibly because of the pharmacokinetic limitations of these molecules. In the search for tachykinin NK(3) receptor antagonists with improved properties, we have discovered GSK172981 and GSK256471. Both compounds demonstrated high affinity for recombinant human (pK(i) values 7.7 and 8.9, respectively) and native guinea pig (pK(i) values 7.8 and 8.4, respectively) tachykinin NK(3) receptors. In vitro functional evaluations revealed GSK172981 to be a competitive antagonist (pA(2)=7.2) at cloned human tachykinin NK(3) receptor whereas GSK256471 diminished the neurokinin B-induced E(max) response, indicative of non-surmountable antagonist pharmacology (pA(2)=9.2). GSK172981 also exhibited a competitive profile in antagonizing neurokinin B-stimulated neuronal activity recorded from the guinea pig medial habenula slices (apparent pK(B)=8.1), whilst GSK256471 abolished the agonist-induced response. Central nervous system penetration by GSK172981 and GSK256471 was indicated by dose-dependent ex vivo tachykinin NK(3) receptor occupancy in medial prefrontal cortex (ED(50) values of 0.8 and 0.9 mg/kg, i.p., respectively) and the dose-dependent attenuation of agonist-induced "wet dog shake" behaviours in guinea pigs. Finally, in vivo microdialysis studies demonstrated that acute GSK172981 (30 mg/kg, i.p.) and GSK256471 (1mg/kg, i.p.) attenuated haloperidol-induced increases in extracellular dopamine in the guinea pig nucleus accumbens. Taken together, these in vitro and in vivo characterisations of the tachykinin NK(3) receptor antagonists GSK172981 and GSK256471 support their potential utility in the treatment of schizophrenia.


Subject(s)
Aminoquinolines/metabolism , Aminoquinolines/pharmacology , Brain/drug effects , Brain/metabolism , Quinolines/metabolism , Quinolines/pharmacology , Receptors, Tachykinin/antagonists & inhibitors , Aminoquinolines/pharmacokinetics , Animals , Behavior, Animal/drug effects , Brain/cytology , Brain/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cloning, Molecular , Dopamine/metabolism , Extracellular Space/drug effects , Extracellular Space/metabolism , Guinea Pigs , Habenula/cytology , Haloperidol/pharmacology , Humans , Inositol Phosphates/metabolism , Male , Microdialysis , Neurokinin B/pharmacology , Neurons/cytology , Neurons/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Peptide Fragments/pharmacology , Peptides/chemistry , Quinolines/pharmacokinetics , Rats , Receptors, Tachykinin/genetics , Receptors, Tachykinin/metabolism , Substance P/analogs & derivatives , Substance P/pharmacology
8.
Clin J Sport Med ; 19(6): 482-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19898076

ABSTRACT

OBJECTIVE: To determine the accuracy of clinically diagnosed exercise-induced bronchospasm (EIB) and asthma among National Collegiate Athletic Association Division I student athletes. DESIGN: Cohort study. SETTING: Division I university athletic department and primary care sports medicine clinic/athletic training room. PATIENTS: All varsity athletes were eligible for the study. Seventy-five participants entered the study; 74 completed all the study protocols. INTERVENTIONS: Study participants underwent measurement of exhaled nitric oxide (eNO), followed by baseline spirometry. Eucapnic voluntary hyperventilation (EVH) was then performed, followed by spirometry every 3 minutes for a total of 21 minutes to measure change in forced expiratory volume in 1 second. MAIN OUTCOME MEASURES: Exhaled nitric oxide levels, baseline spirometry, and response of forced expiratory volume in 1 second to EVH. RESULTS: There were a total of 16 subjects with a positive EVH test. There were 16 (80%) individuals using a bronchodilator who had a negative EVH test versus 4 (20%) who had a positive test. Mean eNO values were different across subject groups defined by treatment status, although not statistically significant. The highest mean eNO value of 24.32 ppb was found in subjects only taking bronchodilators compared with the lowest mean value of 16.75 ppb in athletes who were not taking any medications. CONCLUSIONS: The results from this study provide further evidence to support the need for objective testing to correctly diagnose EIB. These data suggest that measuring eNO may help to distinguish truly isolated EIB from asthma in athletes, but a larger study is needed to confirm these initial observations.


Subject(s)
Asthma, Exercise-Induced/diagnosis , Athletes , Adolescent , Asthma/diagnosis , Breath Tests , Case-Control Studies , Forced Expiratory Volume , Humans , Male , Nitric Oxide/analysis , Spirometry , Young Adult
9.
Eur Neuropsychopharmacol ; 19(11): 778-90, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19584022

ABSTRACT

Social isolation from weaning in rats produces behavioural and hippocampal structural changes at adulthood. Here, rats were group or isolation reared for eight-weeks. Following the initial four-week period of rearing, fluoxetine (10 mg/kg i.p.) was administered for 28 days. Changes in recognition memory, hippocampal monoamines, and cytoskeletal microtubules were investigated. Isolation-rearing for four- or eight-weeks produced recognition memory deficits that were not reversed by fluoxetine. Eight-weeks of isolation decreased alpha-tubulin acetylation (Acet-Tub) and the tyrosinated/detyrosinated alpha-tubulin ratio (Tyr/Glu-Tub), suggesting major alterations in microtubule dynamics and neuronal plasticity. In grouped rats, fluoxetine decreased Acet-Tub without changes in Tyr/Glu-Tub. In isolates, fluoxetine did not affect Acet-Tub but increased Tyr/Glu-Tub. Finally, fluoxetine altered serotonin metabolism in grouped, but not in isolated animals. Therefore, isolation-rearing changes the hippocampal responses of the serotonergic and microtubular system to fluoxetine. These findings show that early-life experience induces behavioural changes paralleled by alterations in cytoskeletal and neurochemical functions.


Subject(s)
Fluoxetine/pharmacology , Hippocampus , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin/metabolism , Social Isolation , Tubulin/metabolism , Analysis of Variance , Animals , Animals, Newborn , Biogenic Monoamines/metabolism , Body Weight/drug effects , Choice Behavior/drug effects , Chromatography, High Pressure Liquid/methods , Exploratory Behavior/drug effects , Handling, Psychological , Hippocampus/drug effects , Hippocampus/metabolism , Male , Motor Activity/drug effects , Protein Isoforms/metabolism , Rats , Tandem Mass Spectrometry/methods
10.
Neuropsychopharmacology ; 34(9): 2156-66, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19404243

ABSTRACT

Alterations in muscarinic acetylcholine receptor (CHRM) populations have been implicated in the pathology of schizophrenia. Here we have assessed whether the receptor function of the M(1) subtype (CHRM1) is altered in a sub-population of patients with schizophrenia, defined by marked (60-80%) reductions in cortical [3H]-pirenzepine (PZP) binding, and termed 'muscarinic receptor-deficit schizophrenia' (MRDS). Using a [35S]-GTPgammaS-Galpha(q/11) immunocapture method we have assessed whether CHRM1 signalling in human cortex (Brodmann area 9 (BA9)) is altered in post mortem tissue from a MRDS group compared with a subgroup of patients with schizophrenia displaying normal PZP binding, and controls with no known history of psychiatric or neurological disorders. The CHRM agonist (oxotremorine-M) and a CHRM1-selective agonist (AC-42) increased Galpha(q/11)-[35S]-GTPgammaS binding, with AC-42 producing responses that were approximately 50% of those maximally evoked by the full agonist, oxotremorine-M, in control and subgroups of patients with schizophrenia. However, the potency of oxotremorine-M to stimulate Galpha(q/11)-[35S]-GTPgammaS binding was significantly decreased in the MRDS group (pEC(50) (M)=5.69+/-0.16) compared with the control group (6.17+/-0.10) and the non-MRDS group (6.05+/-0.07). The levels of Galpha(q/11) protein present in BA9 did not vary with diagnosis. Maximal oxotremorine-M-stimulated Galpha(q/11)-[35S]-GTPgammaS binding in BA9 membranes was significantly increased in the MRDS group compared with the control group. Similar, though non-statistically significant, trends were observed for AC-42. These data provide evidence that both orthosterically and allosterically acting CHRM agonists can stimulate a receptor-driven functional response ([35S]-GTPgammaS binding to Galpha(q/11)) in membranes prepared from post mortem human dorsolateral prefrontal cortex of patients with schizophrenia and controls . Furthermore, in a subgroup of patients with schizophrenia displaying markedly decreased PZP binding (MRDS) we have shown that although agonist potency may decrease, the efficacy of CHRM1-Galpha(q/11) coupling increases, suggesting an adaptative change in receptor-G protein coupling efficiency in this endophenotype of patients with schizophrenia.


Subject(s)
Prefrontal Cortex/metabolism , Receptors, Muscarinic/metabolism , Schizophrenia/metabolism , Adult , Aged , Cell Membrane/drug effects , Cell Membrane/metabolism , Female , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Male , Middle Aged , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Oxotremorine/analogs & derivatives , Oxotremorine/pharmacology , Phenotype , Piperidines/pharmacology , Pirenzepine/pharmacology , Prefrontal Cortex/drug effects , Protein Binding/drug effects , Receptor, Muscarinic M1 , Schizophrenia/drug therapy , Sulfur Radioisotopes , Tritium , Young Adult
11.
Synapse ; 63(4): 359-64, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19140168

ABSTRACT

A number of studies suggest that stressful conditions can induce structural alterations in the hippocampus and that antidepressant drugs may prevent such deficits. In particular, the selective serotonin reuptake inhibitor (SSRI) fluoxetine was more effective in modulating different neuronal plasticity phenomena and related molecules in rat hippocampus. Cytoskeletal microtubule dynamics are fundamental to dendrites and axons remodeling, leading to the hypothesis that fluoxetine may affect the microtubular system. However, despite reports of stress-induced alterations in microtubule dynamics by different stressors, only few studies investigated the in vivo effects of antidepressants on microtubules in specific rat brain regions. The present study investigated the dose-related (1, 5, or 10 mg/kg i.p.) effects of acute and chronic (21 days) treatments with fluoxetine on the ratio of hippocampal alpha-tubulin isoforms which is thought to reflect microtubule dynamics. Western Blot analysis was used to quantify alpha-tubulin isoforms, high-performance liquid chromatography and fluorescence detection was used to measure ex vivo monoamine metabolism. The results showed that acute fluoxetine increased the stable forms acetylated and detyrosinated alpha-tubulin. Conversely, chronic fluoxetine decreased acetylated alpha-tubulin, indicative of increased microtubule dynamics. The neuron-specific Delta2-Tubulin was increased by chronic fluoxetine indicating neuronal involvement in the observed cytoskeletal changes. Although acute and chronic fluoxetine similarly altered serotonin metabolism by inhibition of serotonin reuptake, this showed no apparent correlation to the cytoskeletal perturbations. Our findings demonstrate that fluoxetine administration modulates microtubule dynamics in rat hippocampus. The cytoskeletal effect exerted by fluoxetine may eventually culminate in promoting events of structural neuronal remodeling.


Subject(s)
Cytoskeleton/metabolism , Fluoxetine/administration & dosage , Hippocampus/drug effects , Microtubules/metabolism , Selective Serotonin Reuptake Inhibitors/administration & dosage , Animals , Dose-Response Relationship, Drug , Male , Rats , Time Factors
12.
Neuropharmacology ; 54(8): 1166-74, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18455201

ABSTRACT

While there is now substantial evidence that 5-HT(6) antagonism leads to significantly improved cognitive ability, the mechanism(s) and/or pathway(s) involved are poorly understood. We have evaluated the consequence of chronic administration of the 5-HT(6) receptor antagonists SB-271046 and SB-399885 on neural cell adhesion molecule polysialylation state (NCAM PSA), a neuroplastic mechanism necessary for memory consolidation. Quantitative analysis of NCAM PSA immunopositive neurons in the dentate gyrus of drug-treated animals revealed a dose-dependent increase in polysialylated cell frequency following treatment with both SB-271046 and SB-399885. These effects could not be attributed to increased neurogenesis, as no difference in the rate of bromodeoxyuridine incorporation was apparent between the control and drug-treated groups. A substantial increase in the frequency of polysialylated cells in layer II of the entorhinal and perirhinal cortices was also observed, brain regions not previously associated with neurogenesis. Chronic treatment with SB-271046 or SB-399885 also significantly increased the activation of dentate polysialylation that is specific to learning. This effect does not occur with other cognition-enhancing drugs, such as tacrine, and this action potentially differentiates 5-HT(6) receptor antagonism as an unique neuroplastic mechanism for cognitive processes which may slow or reverse age/neurodegenerative related memory deficits.


Subject(s)
Dentate Gyrus/drug effects , Hippocampus/drug effects , Neural Cell Adhesion Molecules/pharmacology , Neurons/metabolism , Piperazines/pharmacology , Receptors, Serotonin/drug effects , Serotonin Antagonists/pharmacology , Sialic Acids/pharmacology , Sulfonamides/pharmacology , Thiophenes/pharmacology , Animals , Antimetabolites , Bromodeoxyuridine , Cell Proliferation/drug effects , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Dentate Gyrus/cytology , Dose-Response Relationship, Drug , Entorhinal Cortex/cytology , Entorhinal Cortex/drug effects , Hippocampus/cytology , Immunohistochemistry , Male , Maze Learning/drug effects , Neurons/drug effects , Rats , Rats, Wistar
13.
J Pharmacol Exp Ther ; 325(3): 869-74, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18322150

ABSTRACT

Using a selective Galpha(q/11) protein antibody capture guanosine 5'-O-(3-[35S]thio)triphosphate ([35S]GTPgammaS) binding approach, it has been possible to perform a quantitative pharmacological examination of the functional activity of the M(1) muscarinic acetylcholine receptor (mAChR) in membranes prepared from human postmortem cerebral cortex. Oxotremorine-M caused a > or = 2-fold increase in [35S]GTPgammaS-Galpha(q/11) binding with a pEC(50) of 6.06 +/- 0.16 in Brodmann's areas 23 and 25 that was almost completely inhibited by preincubation of membranes with the M(1) mAChR subtype-selective antagonist muscarinic toxin-7. In addition, the orthosteric and allosteric agonists, xanomeline [3(3-hexyloxy-1,2,5-thiadiazol-4-yl)-1,2,5,6-tetrahydro-1-methylpyridine] and AC-42 (4-n-butyl-1-[4-(2-methylphenyl)-4-oxo-1-butyl]-piperidine hydrogen chloride), increased [35S]-GTPgammaS-Galpha(q/11) binding, but with reduced intrinsic activities, inducing maximal responses that were 42 +/- 1 and 44 +/- 2% of the oxotremorine-M-induced response, respectively. These data indicate that the M(1) receptor is the predominant mAChR subtype coupling to the Galpha(q/11) G protein in these brain regions and that it is possible to quantify the potency and intrinsic activity of full and partial M(1) mAChR receptor agonists in postmortem human brain using a selective Galpha(q/11) protein antibody capture [35S]GTPgammaS binding assay.


Subject(s)
Cerebral Cortex/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Membranes/metabolism , Receptor, Muscarinic M1/metabolism , Aged , Animals , Antibodies/metabolism , Atropine/metabolism , Elapid Venoms/metabolism , Female , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Muscarinic Agonists/metabolism , Muscarinic Antagonists/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M1/agonists , Receptor, Muscarinic M1/antagonists & inhibitors , Sulfur Radioisotopes
14.
Neuropsychopharmacology ; 33(7): 1642-52, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17728699

ABSTRACT

Neurokinin-3 (NK3) receptors are concentrated in forebrain and basal ganglia structures within the mammalian CNS. This distribution, together with the modulatory influence of NK3 receptors on monoaminergic neurotransmission, has led to the hypothesis that NK3 receptor antagonists may have therapeutic efficacy in the treatment of psychiatric disorders. Here we describe the in vitro and in vivo characterization of the highly selective NK3 receptor antagonist talnetant (SB-223412). Talnetant has high affinity for recombinant human NK3 receptors (pKi 8.7) and demonstrates selectivity over other neurokinin receptors (pKi NK2 = 6.6 and NK1<4). In native tissue-binding studies, talnetant displayed high affinity for the guinea pig NK3 receptor (pKi 8.5). Functionally, talnetant competitively antagonized neurokinin B (NKB)-induced responses at the human recombinant receptor in both calcium and phosphoinositol second messenger assay systems (pA2 of 8.1 and 7.7, respectively). In guinea pig brain slices, talnetant antagonized NKB-induced increases in neuronal firing in the medial habenula (pKB = 7.9) and senktide-induced increases in neuronal firing in the substantia nigra pars compacta (pKB = 7.7) with no diminution of maximal agonist efficacy, suggesting competitive antagonism at native NK3 receptors. Talnetant (3-30 mg/kg i.p.) significantly attenuated senktide-induced 'wet dog shake' behaviors in the guinea pig in a dose-dependent manner. Microdialysis studies demonstrated that acute administration of talnetant (30 mg/kg i.p.) produced significant increases in extracellular dopamine and norepinephrine in the medial prefrontal cortex and attenuated haloperidol-induced increases in nucleus accumbens dopamine levels in the freely moving guinea pigs. Taken together, these data demonstrate that talnetant is a selective, competitive, brain-penetrant NK3 receptor antagonist with the ability to modulate mesolimbic and mesocortical dopaminergic neurotransmission and hence support its potential therapeutic utility in the treatment of schizophrenia.


Subject(s)
Brain/drug effects , Quinolines/pharmacology , Receptors, Neurokinin-3/antagonists & inhibitors , Animals , Brain/cytology , Calcium/metabolism , Cell Line, Transformed , Cell Line, Tumor , Dopamine/metabolism , Dose-Response Relationship, Drug , Guinea Pigs , Humans , In Vitro Techniques , Male , Neurons/drug effects , Neurotransmitter Agents/metabolism , Protein Binding/drug effects , Receptors, Neurokinin-3/drug effects , Receptors, Neurokinin-3/metabolism
15.
J Med Chem ; 50(21): 5076-89, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17867665

ABSTRACT

The discovery of new highly potent and selective dopamine D3 receptor antagonists has recently permitted characterization of the role of the dopamine D3 receptor in a wide range of preclinical animal models. A novel series of 1,2,4-triazol-3-yl-thiopropyl-tetrahydrobenzazepines demonstrating a high level of D3 affinity and selectivity with an excellent pharmacokinetic profile is reported here. In particular, the pyrazolyl derivative 35 showed good oral bioavailability and brain penetration associated with high potency and selectivity in vitro. In vivo characterization of 35 confirmed that this compound blocks the expression of nicotine- and cocaine-conditioned place preference in the rat, prevents nicotine-triggered reinstatement of nicotine-seeking behavior in the rat, reduces oral operant alcohol self-administration in the mouse, increases extracellular levels of acetylcholine in the rat medial prefrontal cortex, and potentiates the amplitude of the relative cerebral blood volume response to d-amphetamine in a regionally specific manner in the rat brain.


Subject(s)
Benzazepines/chemical synthesis , Receptors, Dopamine D3/antagonists & inhibitors , Triazoles/chemical synthesis , Acetylcholine/metabolism , Administration, Oral , Alcohol Drinking/prevention & control , Animals , Benzazepines/pharmacokinetics , Benzazepines/pharmacology , Brain/blood supply , Brain/metabolism , Cocaine/pharmacology , Conditioning, Operant/drug effects , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/metabolism , Guinea Pigs , Histamine H1 Antagonists/chemical synthesis , Histamine H1 Antagonists/pharmacokinetics , Histamine H1 Antagonists/pharmacology , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Molecular , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D3/agonists , Receptors, Histamine H1/metabolism , Structure-Activity Relationship , Tobacco Use Disorder/prevention & control , Triazoles/pharmacokinetics , Triazoles/pharmacology
16.
Neuropsychopharmacology ; 32(10): 2163-72, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17356576

ABSTRACT

Preclinically, the combination of an SSRI and 5-HT autoreceptor antagonist has been shown to reduce the time to onset of anxiolytic activity compared to an SSRI alone. In accordance with this, clinical data suggest the coadministration of an SSRI and (+/-) pindolol can decrease the time to onset of anxiolytic/antidepressant activity. Thus, the dual-acting novel SSRI and 5-HT(1A/B) receptor antagonist, SB-649915-B, has been assessed in acute and chronic preclinical models of anxiolysis. SB-649915-B (0.1-1.0 mg/kg, i.p.) significantly reduced ultrasonic vocalization in male rat pups separated from their mothers (ED(50) of 0.17 mg/kg). In the marmoset human threat test SB-649915-B (3.0 and 10 mg/kg, s.c.) significantly reduced the number of postures with no effect on locomotion. In the rat high light social interaction (SI), SB-649915-B (1.0-7.5 mg/kg, t.i.d.) and paroxetine (3.0 mg/kg, once daily) were orally administered for 4, 7, and 21 days. Ex vivo inhibition of [(3)H]5-HT uptake was also measured following SI. SB-649915-B and paroxetine had no effect on SI after 4 days. In contrast to paroxetine, SB-649915-B (1.0 and 3.0 mg/kg, p.o., t.i.d.) significantly (p<0.05) increased SI time with no effect on locomotion, indicative of an anxiolytic-like profile on day 7. Anxiolysis was maintained after chronic (21 days) administration by which time paroxetine also increased SI significantly. 5-HT uptake was inhibited by SB-649915-B at all time points to a similar magnitude as that seen with paroxetine. In conclusion, SB-649915-B is acutely anxiolytic and reduces the latency to onset of anxiolytic behavior compared to paroxetine in the SI model.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety Disorders/drug therapy , Benzoxazines/pharmacology , Brain/drug effects , Piperidines/pharmacology , Quinolines/pharmacology , Receptor, Serotonin, 5-HT1A/drug effects , Serotonin/metabolism , Animals , Anxiety Disorders/metabolism , Anxiety Disorders/physiopathology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/metabolism , Brain/physiopathology , Callithrix , Drug Synergism , Male , Motor Activity/drug effects , Motor Activity/physiology , Paroxetine/pharmacology , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Serotonin 5-HT1 Receptor Agonists , Serotonin Antagonists/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Social Behavior , Vocalization, Animal/drug effects , Vocalization, Animal/physiology
17.
Psychopharmacology (Berl) ; 192(1): 121-33, 2007 May.
Article in English | MEDLINE | ID: mdl-17265079

ABSTRACT

RATIONALE: The delay in onset and treatment resistance of subpopulations of depressed patients to conventional serotonin reuptake inhibitors has lead to new drug development strategies to produce agents with improved antidepressant efficacy. OBJECTIVES: We report the in vivo characterization of the novel 5-HT(1A/1B) autoreceptor antagonist/5-HT transporter inhibitor (6-[(1-{2-[(2-methyl-5-quinolinyl)oxy]ethyl}-4-piperidinyl)methyl]-2H-1,4-benzoxazin-3(4H)-one), SB-649915-B. MATERIALS AND METHODS: Ex vivo binding was used to ascertain 5-HT(1A) receptor and serotonin transporter occupancy. 8-OH-DPAT-induced hyperlocomotion and SKF-99101-induced elevation of seizure threshold were used as markers of central blockade of 5-HT(1A) and 5-HT(1B) receptors, respectively. In vivo electrophysiology in the rat dorsal raphe and microdialysis in freely moving guinea pigs and rats were used to evaluate the functional outcome of SB-649915-B. RESULTS: SB-649915-B (1-10 mg/kg p.o.) produced a dose-related inhibition of 5-HT(1A) receptor radioligand binding and inhibited ex vivo [(3)H]5-HT uptake in both guinea pig and rat cortex. SB-649915-B (0.1-10 mg/kg p.o.) reversed both 8-OH-DPAT-induced hyperlocomotor activity and SKF-99101-induced elevation of seizure threshold in the rat, demonstrating in vivo blockade of both 5-HT(1A) and 5-HT(1B) receptors, respectively. SB-649915-B (0.1-3 mg/kg i.v.) produced no change in raphe 5-HT neuronal cell firing per se but attenuated the inhibitory effect of 8-OH-DPAT. Acute administration of SB-649915-B resulted in increases (approximately two- to threefold) in extracellular 5-HT in the cortex of rats and the dentate gyrus and cortex of guinea pigs. CONCLUSIONS: Based on these data, one may speculate that the 5-HT autoreceptor antagonist/5-HT transport inhibitor SB-649915-B will have therapeutic efficacy in the treatment of affective disorders with the potential for a faster onset of action compared to current selective serotonin reuptake inhibitors.


Subject(s)
Benzoxazines/pharmacology , Piperidines/pharmacology , Quinolines/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin 5-HT1 Receptor Antagonists , Serotonin Plasma Membrane Transport Proteins/drug effects , Animals , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Dose-Response Relationship, Drug , Electrophysiology , Electroshock , Guinea Pigs , Locomotion/drug effects , Male , Microdialysis , Mood Disorders/drug therapy , Neurons/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Radioligand Assay , Raphe Nuclei , Rats , Rats, Sprague-Dawley , Seizures , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Species Specificity
18.
Bioorg Med Chem Lett ; 17(2): 400-5, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17084080

ABSTRACT

A rational structure-activity relationship study around compound (1) is reported. The lead optimisation programme led to the identification of sulfonamide (25), a molecule combining dopamine D2/D3 receptor antagonism with serotonin 5-HT2A, 5-HT2C, 5-HT6 receptor antagonism for an effective treatment of schizophrenia. Compound (25) was shown to possess the required in vivo activity with no EPS liability.


Subject(s)
Antipsychotic Agents/chemical synthesis , Antipsychotic Agents/pharmacology , Alkylation , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Dopamine Antagonists/chemical synthesis , Dopamine Antagonists/pharmacology , Dopamine D2 Receptor Antagonists , Drug Design , Humans , Receptor, Serotonin, 5-HT2A/drug effects , Receptor, Serotonin, 5-HT2C/drug effects , Receptors, Dopamine D3/antagonists & inhibitors , Receptors, Serotonin/drug effects , Recombinant Proteins/drug effects , Serotonin Antagonists/chemical synthesis , Serotonin Antagonists/pharmacology , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology
19.
Neuropharmacology ; 51(3): 566-77, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16846620

ABSTRACT

This study utilised the selective 5-ht(5A) receptor antagonist, SB-699551-A (3-cyclopentyl-N-[2-(dimethylamino)ethyl]-N-[(4'-{[(2-phenylethyl)amino]methyl}-4-biphenylyl)methyl]propanamide dihydrochloride), to investigate 5-ht5A receptor function in guinea pig brain. SB-699551-A competitively antagonised 5-HT-stimulated [35S]GTPgammaS binding to membranes from human embryonic kidney (HEK293) cells transiently expressing the guinea pig 5-ht5A receptor (pA2 8.1+/-0.1) and displayed 100-fold selectivity versus the serotonin transporter and those 5-HT receptor subtypes (5-HT(1A/B/D), 5-HT2A/C and 5-HT7) reported to modulate central 5-HT neurotransmission in the guinea pig. In guinea pig dorsal raphe slices, SB-699551-A (1 microM) did not alter neuronal firing per se but attenuated the 5-CT-induced depression in serotonergic neuronal firing in a subpopulation of cells insensitive to the 5-HT1A receptor-selective antagonist WAY-100635 (100 nM). In contrast, SB-699551-A (100 or 300 nM) failed to affect both electrically-evoked 5-HT release and 5-CT-induced inhibition of evoked release measured using fast cyclic voltammetry in vitro. SB-699551-A (0.3, 1 and 3 mg/kg s.c.) did not modulate extracellular levels of 5-HT in the guinea pig frontal cortex in vivo. However, when administered in combination with WAY-100635 (0.3 mg/kg s.c.), SB-699551-A (0.3, 1 or 3 mg/kg s.c.) produced a significant increase in extracellular 5-HT levels. These studies provide evidence for an autoreceptor role for the 5-ht5A receptor in guinea pig brain.


Subject(s)
Neurons/drug effects , Receptors, Serotonin/physiology , Serotonin Antagonists/pharmacology , Serotonin/metabolism , Analysis of Variance , Animals , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacokinetics , Biphenyl Compounds/pharmacology , Brain/cytology , Brain/drug effects , Cell Line , Citalopram/pharmacokinetics , Dose-Response Relationship, Drug , Electrochemistry/methods , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Guinea Pigs , Humans , Isotopes/pharmacokinetics , Lysergic Acid Diethylamide/pharmacokinetics , Male , Microdialysis/methods , Piperazines/pharmacology , Protein Binding/drug effects , Pyridines/pharmacology , Radioligand Assay/methods , Serotonin Antagonists/chemistry , Serotonin Antagonists/pharmacokinetics , Serotonin Receptor Agonists/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacokinetics
20.
Brain Res ; 1097(1): 123-32, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16730678

ABSTRACT

Neonatal maternal separation (MS) has been used to model long-term changes in neurochemistry and behaviour associated with exposure to early-life stress. This study characterises changes in behavioural and neuroendocrine parameters following MS. On postnatal days (PND) 3-15, male and female Long-Evans rats underwent 3 h daily MS. Non-handled (NH) control offspring remained with the dams. Starting at PND 90, behaviour was assessed at weekly intervals in the elevated plus-maze, elevated T-maze, and locomotor activity boxes, and body weight monitored throughout. At the end of the study, adrenals were weighed and blood collected for analysis of plasma corticosterone and adrenocorticotropic hormone (ACTH) under basal conditions and following restraint stress. As adults, MS weighed more than NH animals. Activity on the open arms of the plus-maze was similar between MS and NH animals. In the T-maze, MS males had shorter emergence latencies than their NH counterparts. Spontaneous ambulation in a novel environment was significantly higher in MS than in NH animals, and males exhibited overall lower activity than females. Basal plasma corticosterone was lower in MS than in NH females, but no rearing condition difference was observed following restraint stress. Females had higher corticosterone and ACTH levels than males, whereas adrenal glands of MS animals weighed less than those of NH controls. The MS paradigm caused long-term gender dependent effects on behaviour and HPA axis status. The consistent gender differences confirm and expand existing results showing altered anxiety and stress reactivity in male and female rats.


Subject(s)
Adrenocorticotropic Hormone/blood , Corticosterone/blood , Maternal Deprivation , Maze Learning/physiology , Motor Activity/physiology , Sex Characteristics , Animals , Animals, Newborn , Female , Male , Neurosecretory Systems/metabolism , Rats , Rats, Long-Evans , Time
SELECTION OF CITATIONS
SEARCH DETAIL