Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
2.
NPJ Parkinsons Dis ; 9(1): 114, 2023 Jul 17.
Article in English | MEDLINE | ID: mdl-37460603

ABSTRACT

Direct targeting of alpha-synuclein (ASYN) has emerged as a disease-modifying strategy for Parkinson's disease and other synucleinopathies which is being approached using both small molecule compounds and ASYN-targeted biologics. Minzasolmin (UCB0599) is an orally bioavailable and brain-penetrant small molecule ASYN misfolding inhibitor in clinical development as a disease-modifying therapeutic for Parkinson's disease. Herein the results of preclinical evaluations of minzasolmin that formed the basis for subsequent clinical development are described. Pharmacokinetic evaluations of intraperitoneal 1 and 5 mg/kg minzasolmin in wildtype mice revealed parallel and dose-proportional exposures in brain and plasma. Three-month administration studies in the Line 61 transgenic mouse model of PD were conducted to measure ASYN pathology and other PD-relevant endpoints including markers of CNS inflammation, striatal DAT labeling and gait. Reductions in ASYN pathology were correlated with improved aspects of gait and balance, reductions in CNS inflammation marker abundance, and normalized striatal DAT levels. These findings provide support for human dose determinations and have informed the translational strategy for clinical trial design and biomarker selection for the ongoing clinical studies of minzasolmin in patients living with early-stage Parkinson's disease (ClinicalTrials.gov ID: NCT04658186; EudraCT Number 2020-003265).

3.
Int J Inflam ; 2022: 2337363, 2022.
Article in English | MEDLINE | ID: mdl-35265316

ABSTRACT

Toll-like receptors (TLRs) play a critical role in innate immune system responses to damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs). A growing body of evidence suggests that excessive TLR-mediated innate immune system activation can lead to neuronal damage and precipitate or perpetuate neurodegenerative diseases. Among TLR subtypes, both TLR2 and TLR9 have been implicated in neurodegenerative disorders with increased expression of these receptors in the central nervous system being associated with pro-inflammatory signaling and increased burdens of pathologic aggregated proteins. In the current study, we characterized the actions of a combined TLR2/TLR9 antagonist, NPT1220-312, on pro-inflammatory signaling and cytokine release in monocyte/macrophage-derived heterologous cells, human microglia, and murine and human whole blood. NPT1220-312 potently blocked TLR2- and TLR9-mediated release of inflammatory cytokines in monocyte/macrophage cells and in human microglia. NPT1220-312 also blocked TLR2-mediated activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome including IL-1ß, IL-18, and apoptosis-associated speck-like protein containing a CARD (ASC) release to the culture medium of human differentiated macrophages. The ability of NPT1220-312 to inhibit TLR2 mediated pro-inflammatory release of chemokines and cytokines in situ was demonstrated using murine and human whole blood. Together, these findings suggest that blockade of TLR2 and TLR9 may reduce inappropriate production of pro-inflammatory cytokines and chemokines from peripheral and central immune cells and thus potentially provide therapeutic benefit in neuroinflammatory/neurodegenerative disorders.

4.
Brain ; 144(12): 3692-3709, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34117864

ABSTRACT

NPT520-34 is a clinical stage, small molecule being developed for the treatment of Parkinson's disease and other neurodegenerative disorders. The therapeutic potential of NPT520-34 was first suggested by findings from cell-based assays of alpha-synuclein clearance. As reported here, NPT520-34 was subsequently evaluated for therapeutically relevant actions in a transgenic animal model of Parkinson's disease that overexpresses human alpha-synuclein and in an acute lipopolysaccharide-challenge model using wild-type mice. Daily administration of NPT520-34 to mThy1-alpha-synuclein (Line 61) transgenic mice for 1 or 3 months resulted in reduced alpha-synuclein pathology, reduced expression of markers of neuroinflammation, and improvements in multiple indices of motor function. In a lipopolysaccharide-challenge model using wild-type mice, a single dose of NPT520-34 reduced lipopolysaccharide-evoked increases in the expression of several pro-inflammatory cytokines in plasma. These findings demonstrate the beneficial effects of NPT520-34 on both inflammation and protein-pathology end points, with consequent improvements in motor function in an animal model of Parkinson's disease. These findings further indicate that NPT520-34 may have two complementary actions: (i) to increase the clearance of neurotoxic protein aggregates; and (ii) to directly attenuate inflammation. NPT520-34 treatment may thereby address two of the predominate underlying pathophysiological aspects of neurodegenerative disorders such as Parkinson's disease.


Subject(s)
Brain/drug effects , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/pathology , Animals , Brain/pathology , Humans , Mice , Mice, Transgenic , Synucleinopathies/pathology
5.
Bioorg Med Chem Lett ; 40: 127861, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33636302

ABSTRACT

Toll-like receptors (TLRs) play key role in innate immune response to Damage Associated Molecular Patterns (DAMPs) and Pathogen Associated Molecular Patterns (PAMPs). DAMP/PAMP-mediated activation of TLRs triggers NFκB signaling resulting in pro-inflammatory cytokine release. Using TLR2-Pam2CSK4 agonist co-crystal structure information, we designed and synthesized a novel series of Toll-like Receptor 2 (TLR2) lipid antagonists and identified compounds 14, 15 and 17 with sub-micromolar potency. TLR2 antagonists that we identified are stable for > 1.0 h in both gastric juice and PBS buffer and could be used as research tools.


Subject(s)
Lipids/chemistry , Oligopeptides/chemistry , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 9/agonists , Crystallization , Cytokines/metabolism , Drug Discovery , Humans , NF-kappa B/metabolism , Protein Binding , Signal Transduction , Structure-Activity Relationship , Toll-Like Receptor 2/chemistry , Toll-Like Receptor 9/chemistry
6.
ACS Chem Neurosci ; 4(9): 1249-55, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-23898966

ABSTRACT

Drugs that selectively activate estrogen receptor ß (ERß) are potentially safer than the nonselective estrogens currently used in hormonal replacement treatments that activate both ERß and ERα. The selective ERß agonist AC-186 was evaluated in a rat model of Parkinson's disease induced through bilateral 6-hydroxydopamine lesions of the substantia nigra. In this model, AC-186 prevented motor, cognitive, and sensorimotor gating deficits and mitigated the loss of dopamine neurons in the substantia nigra, in males, but not in females. Furthermore, in male rats, 17ß-estradiol, which activates ERß and ERα with equal potency, did not show the same neuroprotective benefits as AC-186. Hence, in addition to a beneficial safety profile for use in both males and females, a selective ERß agonist has a differentiated pharmacological profile compared to 17ß-estradiol in males.


Subject(s)
Cyclohexanes/therapeutic use , Estrogen Receptor beta/agonists , Neuroprotective Agents/therapeutic use , Parkinsonian Disorders/drug therapy , Phenols/therapeutic use , Animals , Corpus Striatum/chemistry , Corpus Striatum/drug effects , Corpus Striatum/pathology , Cyclohexanes/chemistry , Cyclohexanes/pharmacology , Cytokines/analysis , Drug Evaluation, Preclinical , Estradiol/therapeutic use , Exploratory Behavior/drug effects , Female , Male , Molecular Structure , Nerve Tissue Proteins/analysis , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Ovariectomy , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Phenols/chemistry , Phenols/pharmacology , Rats , Rats, Sprague-Dawley , Reflex, Startle/drug effects , Rotarod Performance Test , Sex Factors , Substantia Nigra/chemistry , Substantia Nigra/drug effects , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/analysis
7.
Behav Pharmacol ; 23(4): 426-33, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22750845

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive deterioration in cognitive functioning. Overall, 25-50% of patients with AD also show symptoms of psychosis including hallucinations and delusions. As all available antipsychotic drugs have a 'black-box' warning for use in these patients because of increased mortality, no appropriate treatment for psychotic symptoms in AD currently exists. In the present study, we examined whether selective antagonism of 5-HT(2A) serotonin receptors has antipsychotic-like activity in an animal model of AD. Mice receiving an intracerebroventricular infusion of the amyloid ß(25-35) peptide fragment showed AD-like histopathology and a psychosis-related behavioral phenotype with enhanced responses to the psychostimulants 2,5-dimethoxy-4-iodoamphetamine hydrochloride and amphetamine as well as disrupted prepulse inhibition. Treatment with pimavanserin, a selective serotonin 5-HT(2A) receptor inverse agonist, prevented 2,5-dimethoxy-4-iodoamphetamine hydrochloride-induced head twitches, reversed the augmented locomotor response to amphetamine, and normalized prepulse inhibition in mice with amyloid pathology. These data suggest that an infusion of amyloid ß might induce alterations in serotonergic function that underlie a psychosis-like phenotype that can be normalized by treatment with a 5-HT(2A) inverse agonist. This in turn suggests that 5-HT(2A) inverse agonists, such as pimavanserin, might have therapeutic benefits in the treatment of psychosis in AD patients.


Subject(s)
Antipsychotic Agents/pharmacology , Piperidines/pharmacology , Psychotic Disorders/drug therapy , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Urea/analogs & derivatives , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Amphetamine/pharmacology , Amphetamines/pharmacology , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Drug Inverse Agonism , Male , Mice , Psychotic Disorders/etiology , Urea/pharmacology
8.
Naunyn Schmiedebergs Arch Pharmacol ; 385(3): 313-23, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22083558

ABSTRACT

Current antipsychotic drug therapies for schizophrenia have limited efficacy and are notably ineffective at addressing the cognitive deficits associated with this disorder. The present study was designed to develop effective antipsychotic agents that would also ameliorate the cognitive deficits associated with this disease. In vitro studies comprised of binding and functional assays were utilized to identify compounds with the receptor profile that could provide both antipsychotic and pro-cognitive features. Antipsychotic and cognitive models assessing in vivo activity of these compounds included locomotor activity assays and novel object recognition assays. We developed a series of potential antipsychotic agents with a novel receptor activity profile comprised of muscarinic M(1) receptor agonism in addition to dopamine D(2) antagonism and serotonin 5-HT(2A) inverse agonism. Like other antipsychotic agents, these compounds reverse both amphetamine and dizocilpine-induced hyperactivity in animals. In addition, unlike other antipsychotic drugs, these compounds demonstrate pro-cognitive actions in the novel object recognition assay. The dual attributes of antipsychotic and pro-cognitive actions distinguish these compounds from other antipsychotic drugs and suggest that these compounds are prototype molecules in the development of novel pro-cognitive antipsychotic agents.


Subject(s)
Antipsychotic Agents/pharmacology , Cognition/drug effects , Animals , Antipsychotic Agents/chemical synthesis , Antipsychotic Agents/pharmacokinetics , Dopamine D2 Receptor Antagonists , HEK293 Cells , Humans , Male , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Motor Activity/drug effects , NIH 3T3 Cells , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M1/agonists , Receptor, Serotonin, 5-HT2A , Recognition, Psychology/drug effects , Serotonin Antagonists/chemical synthesis , Serotonin Antagonists/pharmacokinetics , Serotonin Antagonists/pharmacology
9.
Behav Pharmacol ; 22(7): 681-92, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21921840

ABSTRACT

Parkinson's disease psychosis (PDP) is a condition for which a safe, tolerated, and effective therapy is lacking. Treatment with typical or atypical antipsychotics may be contraindicated in patients with PDP because of the potential for aggravating motor symptoms. This study used a novel animal model with features of both Parkinson's disease (PD) and psychosis to examine a potential mechanism for reversing PDP. Animals with bilateral 6-hydroxydopamine lesions of the substantia nigra displayed motoric impairments characteristic of humans with PD. In addition, they displayed augmented head twitches, augmented amphetamine-induced locomotor activity, and disrupted prepulse inhibition compared with sham controls, behavioral indices frequently used to assess antipsychotic activity in animal models. Pimavanserin, a selective 5-HT2A antagonist/inverse agonist, reversed the psychotic-like behavioral deficits, suggesting that nigrostriatal (6-hydroxydopamine) lesions induced alterations in 5-HT2A-mediated signaling. The selective 5-HT2A inverse agonist M100907, but not the selective 5-HT2C inverse agonist SB 252084 paralleled the effects of pimavanserin. Of note, the reversal of psychotic-like behaviors produced by 5-HT2A inverse agonists occurred without disrupting motor behaviors in lesioned subjects, suggesting that 5HT2A antagonism/inverse agonism may be beneficial in the treatment of PDP.


Subject(s)
Antipsychotic Agents/pharmacology , Behavior, Animal , Central Nervous System Stimulants/pharmacology , Dyskinesias/metabolism , Parkinson Disease/drug therapy , Piperidines/pharmacology , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Urea/analogs & derivatives , Amphetamine/pharmacology , Amphetamines/pharmacology , Animals , Antipsychotic Agents/therapeutic use , Antipsychotic Agents/toxicity , Disease Models, Animal , Fenfluramine/pharmacology , Fluorobenzenes/pharmacology , Hyperkinesis , Male , Motor Activity , Oxidopamine/toxicity , Parkinson Disease/complications , Parkinson Disease/etiology , Piperidines/therapeutic use , Piperidines/toxicity , Psychotic Disorders/drug therapy , Psychotic Disorders/physiopathology , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A/physiology , Sensory Gating , Serotonin 5-HT2 Receptor Antagonists/toxicity , Serotonin Agents/pharmacology , Serotonin Receptor Agonists/pharmacology , Substantia Nigra/physiology , Tyrosine 3-Monooxygenase/analysis , Urea/pharmacology , Urea/therapeutic use , Urea/toxicity
10.
Neuropharmacology ; 58(2): 365-73, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19835892

ABSTRACT

The recent discovery of allosteric potentiators and agonists of the muscarinic M(1) receptor represents a significant advance in the muscarinic receptor pharmacology. In the current study we describe the receptor pharmacology and pro-cognitive action of the allosteric agonist AC-260584. Using in vitro cell-based assays with cell proliferation, phosphatidylinositol hydrolysis or calcium mobilization as endpoints, AC-260584 was found to be a potent (pEC(50) 7.6-7.7) and efficacious (90-98% of carbachol) muscarinic M(1) receptor agonist. Furthermore, as compared to orthosteric binding agonists, AC-260584 showed functional selectivity for the M(1) receptor over the M(2), M(3), M(4) and M(5) muscarinic receptor subtypes. Using GTPgammaS binding assays, its selectivity was found to be similar in native tissues expressing mAChRs to its profile in recombinant systems. In rodents, AC-260584 activated extracellular signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation in the hippocampus, prefrontal cortex and perirhinal cortex. The ERK1/2 activation was dependent upon muscarinic M(1) receptor activation since it was not observed in M(1) knockout mice. AC-260584 also improved the cognitive performance of mice in the novel object recognition assay and its action is blocked by the muscarinic receptor antagonist pirenzepine. Taken together these results indicate for the first time that a M(1) receptor agonist selective over the other mAChR subtypes can have a symptomatically pro-cognitive action. In addition, AC-260584 was found to be orally bioavailable in rodents. Therefore, AC-260584 may serve as a lead compound in the development of M(1) selective drugs for the treatment of cognitive impairment associated with schizophrenia and Alzheimer's disease.


Subject(s)
Benzoxazines/pharmacology , Cognition/drug effects , Nootropic Agents/pharmacology , Receptor, Muscarinic M1/agonists , Administration, Oral , Animals , Benzoxazines/administration & dosage , Benzoxazines/pharmacokinetics , Biological Availability , Brain/drug effects , Brain/metabolism , CHO Cells , Cognition/physiology , Cricetinae , Cricetulus , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscarinic Agonists/administration & dosage , Muscarinic Agonists/pharmacokinetics , Muscarinic Agonists/pharmacology , NIH 3T3 Cells , Nootropic Agents/administration & dosage , Nootropic Agents/pharmacokinetics , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M1/genetics , Receptor, Muscarinic M1/metabolism , Receptors, Muscarinic/genetics , Receptors, Muscarinic/metabolism , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Spinal Cord/drug effects , Spinal Cord/metabolism
11.
BMC Pharmacol ; 9: 14, 2009 Dec 02.
Article in English | MEDLINE | ID: mdl-19951444

ABSTRACT

BACKGROUND: Activation of muscarinic M1 receptors is mediated via interaction of orthosteric agonists with the acetylcholine binding site or via interaction of allosteric agonists with different site(s) on the receptor. The focus of the present study was to determine if M1 receptors activated by allosteric agonists undergo the same regulatory fate as M1 receptors activated by orthosteric agonists. RESULTS: The orthosteric agonists carbachol, oxotremorine-M and pilocarpine were compared to the allosteric agonists AC-42, AC-260584, N-desmethylclozapine and xanomeline. All ligands activated M1 receptors and stimulated interaction of the receptors with beta-arrestin-1. All ligands reduced cell surface binding and induced the loss of total receptor binding. Receptor internalization was blocked by treatment with hypertonic sucrose indicating that all ligands induced formation of clathrin coated vesicles. However, internalized receptors recycled to the cell surface following removal of orthosteric, but not allosteric agonists. Whereas all ligands induced loss of cell surface receptor binding, no intracellular vesicles could be observed after treatment with AC-260584 or xanomeline. Brief stimulation of M1 receptors with AC-260584 or xanomeline resulted in persistent activation of M1 receptors, suggesting that continual receptor signaling might impede or delay receptor endocytosis into intracellular vesicles. CONCLUSION: These results indicate that allosteric agonists differ from orthosteric ligands and among each other in their ability to induce different regulatory pathways. Thus, signaling and regulatory pathways induced by different allosteric ligands are ligand specific.


Subject(s)
Muscarinic Agonists/chemistry , Muscarinic Agonists/metabolism , Receptor, Muscarinic M1/agonists , Receptor, Muscarinic M1/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Humans , Ligands , Muscarinic Agonists/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Receptor, Muscarinic M1/physiology
12.
Eur J Pharmacol ; 572(2-3): 129-37, 2007 Oct 31.
Article in English | MEDLINE | ID: mdl-17628522

ABSTRACT

Both muscarinic and nicotinic receptors are implicated in cognition. We have previously suggested that stimulation of the muscarinic M1 receptor has a beneficial effect on cognition, based upon evidence that the muscarinic M1 receptor agonist of N-desmethylclozapine, the major metabolite of clozapine, may contribute to the ability of clozapine to improve some domains of cognition in schizophrenia. Present study examined the effectiveness of a new muscarinic M1 receptor agonist, 4-[3-(4-butylpiperidin-1-yl)-propyl]-7-fluoro-4H-benzo[1,4]oxazin-3-one (AC260584), to increase the release of acetylcholine and dopamine in the rat medial prefrontal cortex and hippocampus. Using microdialysis in awake, freely moving rats, AC260584, 3 and 10, but not 1 mg/kg (s.c.), significantly increased dopamine release in the medial prefrontal cortex and hippocampus. However, only the high dose of AC260584, 10 mg/kg (s.c.), significantly increased acetylcholine release in these regions. Moreover, the increases in acetylcholine release produced by AC260584, 10 mg/kg, were attenuated by the muscarinic M1 receptor antagonist telenzepine (3 mg/kg, s.c.) but not by the 5-HT1A receptor antagonist N-[2-(4-2-methoxyphenyl)-1-piperazinyl]-N-(2-pyridyl) cyclohexanecarboxamide (WAY100635, 0.2 mg/kg, s.c.). However, the increase in dopamine release produced by 10 mg/kg AC260584 was blocked by both telenzepine and WAY100635. In addition, pretreatment with the atypical antipsychotic drug risperidone (0.1 mg/kg, s.c.) potentiated AC260584 (1.0 mg/kg, s.c.)-induced acetylcholine and dopamine release in the medial prefrontal cortex. These findings suggest that the muscarinic M1 receptor agonist property of AC260584 contributes to its enhancement of cortical acetylcholine and dopamine efflux. Therefore, AC260584, as well as other muscarinic M1 receptor agonists, may be a valuable target for the development of drugs which can improve the cognitive deficits in schizophrenia and perhaps other neuropsychiatric disorders, as well.


Subject(s)
Acetylcholine/metabolism , Antipsychotic Agents/pharmacology , Benzoxazines/pharmacology , Dopamine/metabolism , Hippocampus/drug effects , Prefrontal Cortex/drug effects , Receptor, Muscarinic M1/agonists , Animals , Drug Synergism , Hippocampus/metabolism , Male , Microdialysis , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Risperidone/pharmacology
13.
Pharmacol Ther ; 115(2): 223-31, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17583355

ABSTRACT

Currently available treatments for schizophrenia have limited efficacy and are generally poorly tolerated. However, among these antipsychotic agents, clozapine stands apart in having generally superior motoric tolerability and efficacy. One intriguing possibility, based on clinical correlations, receptor activity profiles and studies with animal models predictive of antipsychotic or cognitive action is that the activity of N-desmethylclozapine (NDMC), a major metabolite of clozapine, may, at least in part, underlie the unique efficacy of clozapine. In this review we compare the pharmacological properties of NDMC to those of clozapine and consider how they may contribute to the overall clinical properties of clozapine. We also consider whether NDMC, in its own right, might be a superior antipsychotic drug.


Subject(s)
Antipsychotic Agents/pharmacology , Clozapine/analogs & derivatives , Clozapine/pharmacology , Animals , Antipsychotic Agents/metabolism , Antipsychotic Agents/therapeutic use , Clozapine/metabolism , Clozapine/therapeutic use , Humans , Receptors, Adrenergic, alpha/drug effects , Receptors, Adrenergic, alpha/physiology , Receptors, Dopamine/drug effects , Receptors, Dopamine/physiology , Receptors, Histamine/drug effects , Receptors, Histamine/physiology , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/physiology , Receptors, Opioid/drug effects , Receptors, Opioid/physiology , Receptors, Serotonin/drug effects , Receptors, Serotonin/physiology , Schizophrenia/drug therapy
14.
J Pharmacol Exp Ther ; 322(2): 862-70, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17519387

ABSTRACT

Dopamine D(2) receptor antagonism contributes to the therapeutic action of antipsychotic drugs (APDs) but also produces undesirable side effects, including extrapyramidal motor deficits, cognitive dulling, and prolactinemia. The introduction of atypical APDs was a significant advancement in the treatment of schizophrenia. Whereas these agents are D(2) receptor antagonists, they are also potent 5-hydroxytryptamine (5-HT)(2A) receptor inverse agonists, a feature that may explain their improved efficacy and tolerability. Recently, we reported that N-(4-fluorophenylmethyl)-N-(1-methylpiperidin-4-yl)-N'-(4-(2-methylpropyloxy)phenylmethyl) carbamide (2R,3R)-dihydroxybutanedioate (2:1) (ACP-103), a novel selective 5-HT(2A) receptor inverse agonist that fails to bind D(2) receptors, is active in several models predictive of antipsychotic activity. Using ACP-103, we tested the hypothesis that combining high levels of 5-HT(2A) inverse agonism with low levels of D(2) antagonism would result in a favorable interaction, such that antipsychotic efficacy could be achieved with reduced D(2) receptor-related adverse effects. Here we show that ACP-103 1) potently inhibited head-twitching produced by the 5-HT(2A/2C) receptor agonist (+/-)-2,5-dimethoxy-4-iodoamphetamine, 2) increased the potency of haloperidol against amphetamine-induced hyperactivity, 3) interacted synergistically with haloperidol or risperidone to suppress hyperactivity induced by the N-methyl-d-aspartate receptor antagonist (5R,10S)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine hydrogen maleate (MK-801), and, by contrast, 4) attenuated haloperido-l- or risperidone-induced prolactinemia. ACP-103 also attenuated catalepsy produced by haloperidol or risperidone. However, the doses that were required for this effect were higher than would be expected for a 5-HT(2A) receptor-mediated mechanism. These data indicate that utilizing ACP-103 as an adjunctive therapy to currently used APDs may result in enhanced antipsychotic efficacy while reducing adverse effects including those attributable to D(2) receptor antagonism.


Subject(s)
Haloperidol/pharmacology , Motor Activity/drug effects , Piperidines/pharmacology , Risperidone/pharmacology , Serotonin 5-HT2 Receptor Agonists , Urea/analogs & derivatives , Amphetamine/pharmacology , Amphetamines/pharmacology , Animals , Antipsychotic Agents/pharmacology , Antipsychotic Agents/toxicity , Behavior, Animal/drug effects , Brain Chemistry , Catalepsy/chemically induced , Catalepsy/prevention & control , Dizocilpine Maleate/pharmacology , Dopamine D2 Receptor Antagonists , Dose-Response Relationship, Drug , Drug Interactions , Drug Synergism , Haloperidol/toxicity , Head Movements/drug effects , Male , Mice , Mice, Inbred Strains , Prolactin/blood , Rats , Rats, Sprague-Dawley , Risperidone/toxicity , Serotonin Receptor Agonists/pharmacology , Urea/pharmacology
15.
Psychopharmacology (Berl) ; 165(2): 128-35, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12404071

ABSTRACT

RATIONALE: Marijuana is known to have psychotropic effects in humans. In this study, we used rat models of sensorimotor gating, hyperactivity and stereotypy to explore whether CB(1) receptor stimulation or blockade induces behavioral changes consistent with psychotomimetic or antipsychotic agents, respectively. OBJECTIVES: We determined whether (a) the cannabinoid agonist CP 55940 decreased pre-pulse inhibition (PPI) as might be expected from a psychotomimetic agent, and (b) the selective CB(1) receptor antagonist, SR 141716A, had any effect on PPI on its own or following disruptions by psychotomimetic agents. In addition, we investigated the effects of SR 141716A on elevated levels of hyperactivity and stereotypy elicited by d-amphetamine. METHODS: These studies were conducted in rats using standard methodologies for determination of PPI following acoustic stimuli, and d-amphetamine-induced hyperactivity and stereotypies. RESULTS: Decreased startle responses to 120 dB stimuli were observed in rats treated with CP 55940 (0.1 mg/kg IP) in the absence and presence of a 73 dB pre-pulse. These effects were reversed by SR 141716A (5 and 10 mg/kg, respectively). SR 141716A (0.1, 5, 10 mg/kg) had no effect on PPI on its own or following disruptions by apomorphine, d-amphetamine or MK-801. Conversely, in separate experiments different antipsychotic agents reversed disruptions in PPI induced by d-amphetamine (haloperidol), apomorphine (haloperidol or clozapine) or MK-801 (clozapine or olanzapine). In addition, unlike haloperidol, SR 141716A (5 mg/kg) did not reverse d-amphetamine-mediated increases in hyperactivity or stereotypy. CONCLUSIONS: The CP 55940-mediated decreases in startle amplitude confound assessment of the effects of CB(1) receptor activation on PPI. The failure of SR 141716A to reverse disruptions in PPI, hyperactivity or stereotypy induced by non-cannabinoid psychotomimetic agents suggests that blockade of the CB(1) receptor on its own is not sufficient for antipsychotic therapy.


Subject(s)
Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Cyclohexanols/pharmacology , Dextroamphetamine/pharmacology , Hallucinogens/pharmacology , Piperidines/pharmacology , Psychoses, Substance-Induced/physiopathology , Pyrazoles/pharmacology , Receptors, Drug/drug effects , Animals , Apomorphine/pharmacology , Arousal/drug effects , Arousal/physiology , Attention/drug effects , Attention/physiology , Behavior, Animal/physiology , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Male , Motor Activity/drug effects , Motor Activity/physiology , Rats , Rats, Wistar , Receptors, Cannabinoid , Receptors, Drug/physiology , Reflex, Startle/drug effects , Rimonabant , Stereotyped Behavior/drug effects , Stereotyped Behavior/physiology
16.
J Biomol Screen ; 7(3): 281-9, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12097191

ABSTRACT

The pharmacology of G protein-coupled receptors is widely accepted to depend on the G protein subunit to which the agonist-stimulated receptor couples. In order to investigate whether CB(1) agonist-mediated signal transduction via an engineered G(alpha 16) system is different than that of the G(i/o) coupling normally preferred by the CB(1) receptor, we transfected the human recombinant CB(1) receptor (hCB(1)) or a fusion protein comprising the hCB(1) receptor and G(alpha 16) (hCB(1)-G(alpha 16)) into HEK293 cells. From competition binding studies, the rank order of ligand affinities at the hCB(1)-G(alpha 16) fusion protein was found to be similar to that for hCB(1): HU 210 > CP 55,940 > or = SR 141716A > WIN 55212-2 > anandamide > JWH 015. Agonists increased [(35)S]GTP gamma S binding or inhibited forskolin-stimulated cAMP, presumably by coupling to G(i/o), in cells expressing hCB(1) but not hCB(1)-G(alpha 16). However, an analogous rank order of potencies was observed for these agonists in their ability to evoke increases in intracellular calcium concentration in cells expressing hCB(1)-G(alpha 16) but not hCB(1). These data demonstrate that ligand affinities for the hCB(1) receptor are not affected by fusion to the G(alpha 16) subunit. Furthermore, there is essentially no difference in the function of the hCB(1) receptor when coupled to G(i/o) or G (alpha 16).


Subject(s)
Heterotrimeric GTP-Binding Proteins/pharmacology , Receptors, Drug/metabolism , Calcium/metabolism , Cyclic AMP/analysis , Cyclohexanols/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11 , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Receptors, Cannabinoid , Recombinant Fusion Proteins/pharmacology , Sulfur Radioisotopes/metabolism , Tritium/metabolism
17.
J Neurosci ; 22(3): 1010-9, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11826129

ABSTRACT

Serotonin is a major component of the inflammatory chemical milieu and contributes to the pain of tissue injury via an action on multiple receptor subtypes. Here we studied mice after genetic or pharmacological disruption of the 5-HT(3) receptor, an excitatory serotonin-gated ion channel. We demonstrate that tissue injury-induced persistent, but not acute, nociception is significantly reduced after functional elimination of this receptor subtype. Specifically, in the setting of tissue injury, the 5-HT(3) receptor mediates activation of nociceptors but does not contribute to injury-associated edema. This result is explained by the localization of 5-HT(3) receptor transcripts to a previously uncharacterized subset of myelinated and unmyelinated afferents, few of which express the proinflammatory neuropeptide substance P. Finally, we provide evidence that central serotonergic circuits modulate nociceptive transmission via a facilitatory action at spinal 5-HT(3) receptors. We conclude that activation of both peripheral and central 5-HT(3) receptors is pronociceptive and that the contribution of peripheral 5-HT(3) receptors involves a novel complement of primary afferent nociceptors.


Subject(s)
Nerve Fibers, Myelinated/metabolism , Nociceptors/metabolism , Pain/metabolism , Receptors, Serotonin/metabolism , Animals , Autoradiography , Behavior, Animal/physiology , Cells, Cultured , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Nerve Fibers/metabolism , Nociceptors/cytology , Pain/genetics , Pain Measurement , Protein Subunits , Radioligand Assay , Receptors, Serotonin/deficiency , Receptors, Serotonin/genetics , Receptors, Serotonin, 5-HT3 , Serotonin/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...