Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters










Publication year range
1.
Behav Brain Res ; 144(1-2): 111-7, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12946601

ABSTRACT

Nociceptin/orphanin FQ (N/OFQ) is a newly discovered neuropeptide that has been implicated in the neurobiological regulation of the behavioral responses to stress and fear. To investigate the role of this peptide in the expression of stress/anxiety-related behaviors in mice, a gene targeting approach to disrupt N/OFQ in the pre-proN/OFQ gene was used. The impact of environmental housing conditions (single and social housing) was assessed on N/OFQ-knockout male and female mice in different experimental paradigms known to trigger distinctive types of stress and anxiety states. Neurological examination of homozygous mutant adult animals indicated that basic neurological functions (vision, audition, olfaction, tactile and pain sensitivity, motor performances) were normal. When housed individually, N/OFQ-knockout animals displayed responses similar to control animals in behavioral tests of emotional reactivity (behavioral despair, locomotor activity, light-dark preference, and acoustic startle tests). In contrast, increased emotional responses were detected when individually housed mice were crowded together (five per cage) under conditions of competitive access to food, water, space, and social contacts. Under those conditions, male mice deficient for N/OFQ developed greater home-cage aggression and increased fear/anxiety-like behaviors in the light-dark and acoustic startle tests, when compared to their wild-type littermates. Group-housed female mutants also showed higher level of anxiety in the acoustic startle test, but needed additional restrain stress to express detectable levels of anxiety in the light-dark test. These data indicate a clear environment-induced rise in fear reactions of N/OFQ-knockout mice. They further suggest that N/OFQ system is essential for development of adequate coping strategies to acute and chronic stress.


Subject(s)
Emotions/physiology , Environment , Gene Deletion , Opioid Peptides/genetics , Analysis of Variance , Animals , Behavior, Animal , Exploratory Behavior/physiology , Light , Mice , Mice, Inbred C57BL , Mice, Knockout/physiology , Mice, Knockout/psychology , Mutation , Neurologic Examination/methods , Opioid Peptides/deficiency , Psychomotor Performance/physiology , Rats , Reflex, Acoustic , Reflex, Startle , Restraint, Physical/methods , Sex , Swimming , Time Factors , Nociceptin
2.
Life Sci ; 70(15): 1719-25, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-12002517

ABSTRACT

Nociceptin/orphanin FQ (N/OFQ) is the endogenous ligand for the opioid receptor-like receptor or nociceptin receptor (NOP). We have compared a novel non-peptide NOP agonist Ro64-6198 with N/OFQ in a series of GTPgamma35S binding and inhibition of forskolin stimulated cAMP formation assays. GTPgamma35S binding assays were performed in membranes prepared from Chinese hamster ovary cells expressing the recombinant human NOP (CHOhNOP). cAMP inhibition studies were performed in whole CHOhNOP cells. Both Ro64-6198 and N/OFQ stimulated GTPgamma35S binding with pEC50 values(95%CL) of 7.61(0.18) and 8.58(0.21) respectively. Both Ro64-6198 and N/OFQ inhibited cAMP formation with pEC50 values of 8.45(0.9) and 9.28(028) respectively. In each assay Ro64-6198 and N/OFQ were full agonists. Ro64-6198 stimulation of GTPgamma35S binding and inhibition of cAMP formation was competitively antagonised by the NOP antagonists [Nphe1]NC(1 - 13)NH2 (10microM), J-113397 (100nM) and III-BTD (1microM) with pKB values of 7.04(0.34) and 6.29(0.10), 8.65(0.34) and 7.90(0.30) and 7.59(0.22) and 7.60(0.22) respectively. Despite the slightly reduced potency of Ro64-6198 compared with N/OFQ, by virtue of high selectivity and relative metabolic stability this molecule will be of considerable use in studies of the actions of the NOP.


Subject(s)
Anti-Anxiety Agents/metabolism , Imidazoles/metabolism , Receptors, Opioid/metabolism , Somatostatin/analogs & derivatives , Spiro Compounds/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Benzimidazoles/metabolism , Benzimidazoles/pharmacology , Binding, Competitive , CHO Cells/drug effects , CHO Cells/metabolism , Cricetinae , Cricetulus , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/antagonists & inhibitors , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Imidazoles/pharmacology , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Narcotic Antagonists , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Piperidines/metabolism , Piperidines/pharmacology , Receptors, Opioid/agonists , Recombinant Fusion Proteins , Somatostatin/metabolism , Somatostatin/pharmacology , Spiro Compounds/pharmacology , Sulfur Radioisotopes , Transfection , Nociceptin Receptor
3.
Eur J Neurosci ; 15(5): 911-22, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11906533

ABSTRACT

Using a combination of the selective opioid receptor-like1 (ORL1) receptor agonist, Ro 64-6198, and orphanin FQ/nociceptin (OFQ/N) peptide knockout (KO) mice, the influence of OFQ/N on cognition has been studied in the rodent. In wild type, C57BL/6J mice, Ro 64-6198 (0.3-1 mg/kg i.p.) impaired the acquisition of spatial learning in the Morris water maze, although a mild neurological impairment was evident which complicated precise interpretation. In Lister hooded rats, Ro 64-6198 (6 mg/kg i.p.) produced delay dependent impairments in rats performing either a delayed matching or a delayed nonmatching to position task with only a modest (< 20%) effect on omissions - an effect consistent with a short-term memory impairment. Electrophysiological studies demonstrated an inhibitory effect of OFQ/N on LTP recorded from the CA1 region of wild type mice, but not in ORL1 receptor knockout mice. In contrast to the ORL1 agonist, mice deficient in the OFQ/N peptide showed some evidence of improved spatial learning, fear conditioning and passive avoidance retention. However, CA1 LTP was similar between OFQ/N peptide KO mice and wild type controls. Subsequent receptor radioautography studies demonstrated the presence of ORL1 receptors within various regions of the medial temporal lobe system: i.e. CA1, dentate gyrus molecular layer, subiculum, perirhinal cortex. Taken together, these results suggest a bi-directional effect of OFQ/N containing systems on aspects of cognitive behaviour, particularly those elements associated with hippocampal function. This is consistent with a likely modulatory role of OFQ/N on hippocampal and associated cortical circuitry.


Subject(s)
Hippocampus/drug effects , Long-Term Potentiation/drug effects , Maze Learning/drug effects , Memory/drug effects , Neurons/drug effects , Opioid Peptides/deficiency , Receptors, Opioid/deficiency , Animals , Anti-Anxiety Agents/pharmacology , Avoidance Learning/drug effects , Avoidance Learning/physiology , Cognition/drug effects , Cognition/physiology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dose-Response Relationship, Drug , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/metabolism , Imidazoles/pharmacology , Long-Term Potentiation/physiology , Male , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Opioid Peptides/genetics , Opioid Peptides/pharmacology , Radioligand Assay , Rats , Rats, Inbred Strains , Receptors, Opioid/agonists , Receptors, Opioid/metabolism , Space Perception/drug effects , Space Perception/physiology , Spiro Compounds/pharmacology , Nociceptin Receptor , Nociceptin
4.
J Recept Signal Transduct Res ; 21(1): 55-70, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11693173

ABSTRACT

We have modified Semliki Forest virus (SFV) vectors to broaden their application range. Here we describe a series of site-directed mutagenesis experiments on the SFV subgenomic 26S promoter to down-regulate the heterologous gene expression. Several mutants showed a dramatic effect on transgene expression levels in BHK cells. The luciferase activity was reduced to approximately 30%, 3%, and 1% compared to the wild type promoter. Similarly, a decrease in beta-galactosidase activity was observed in BHK cells and after injection into the striatum of male Wistar rats. Novel non-cytopathogenic and temperature-sensitive SFV vectors have recently been developed by introduction of point mutations in the viral nonstructural genes nsP2 and nsP4. These vectors do not show the typical shut down of host cell protein synthesis after SFV infections and therefore allow for a substantially prolonged survival of host cells. Both the mutant vectors demonstrating lower and more physiological expression levels and the non-cytopathogenic vectors should be valuable tools for various applications within receptor research. Furthermore, recent studies suggest that SFV vectors can be efficient gene delivery vehicles for gene therapy applications.


Subject(s)
Genetic Vectors , Semliki forest virus/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Brain/metabolism , Brain/virology , Cell Line , Cricetinae , Cytopathogenic Effect, Viral/genetics , DNA, Viral/genetics , Gene Expression , Genes, Reporter , Genetic Therapy , Lac Operon , Luciferases/genetics , Male , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Rats , Rats, Wistar , Receptors, Virus , Research Design , Semliki forest virus/pathogenicity , Temperature , Viral Nonstructural Proteins/genetics
5.
Psychopharmacology (Berl) ; 156(2-3): 273-83, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11549229

ABSTRACT

RATIONALE: Schizophrenic patients typically have impaired startle habituation (SH) and prepulse inhibition of the startle reflex (PPI). PPI can be disrupted in rats by psychomimetics, and drug-induced reversal of this deficit is considered to predict potential antipsychotic properties. Certain strains of mice, such as C57BL/6J, naturally display poor PPI. OBJECTIVE: To test whether mice spontaneously showing low levels of PPI might prove a useful tool for detecting novel antipsychotics. METHODS: PPI and SH were evaluated in four strains of mice: BALB/cByJ, MORO, 129/SvEv and C57BL/6J. The effects of antipsychotic [haloperidol (1, 3 and 6 mg/kg), clozapine (0.3, 1, 3 and 30 mg/kg) and risperidone (0.1, 0.3 and 1 mg/kg)] and non-antipsychotic [diazepam (3, 10 and 30 mg/kg), buspirone (1, 3 and 10 mg/kg), desipramine (3, 10 and 30 mg/kg), morphine (3, 10 and 30 mg/kg) and scopolamine (0.3, 1 and 3 mg/kg)] drug treatments were studied on PPI. RESULTS: Haloperidol (6 mg/kg), clozapine (3 and 30 mg/kg), and risperidone (1 mg/kg) all significantly enhanced PPI in C57BL/6J. All non-antipsychotics failed to improve PPI in this strain, except diazepam. Facilitation of PPI was also obtained in the other strains; however, clear interstrain differences were observed depending on the class of antipsychotic used and on the level of prepulse intensity. CONCLUSION: Antipsychotic-induced facilitation of PPI is clearly detected in mice naturally exhibiting poor levels of sensorimotor gating (e.g., C57BL/6J), but is also observed in other strains of mice. The use of this procedure as a potential screening test for detecting novel antipsychotic medications is discussed.


Subject(s)
Antipsychotic Agents/pharmacology , Reflex, Startle/drug effects , Acoustic Stimulation , Animals , Drug Evaluation, Preclinical , Habituation, Psychophysiologic , Male , Mice , Mice, Inbred Strains , Psychotropic Drugs/pharmacology , Species Specificity , Stimulation, Chemical
6.
J Pharmacol Exp Ther ; 298(2): 812-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11454946

ABSTRACT

The novel nonpeptide orphanin FQ/nociceptin (OFQ/N) ligand [(1S,3aS)-8-(2,3,3a,4,5,6-hexahydro-1H-phenalen-1-yl)-1-phenyl-1,3,8-triaza-spiro[4.5]decan-4-one] (Ro 64-6198) was characterized in vitro and in vivo for its agonistic potential. Ro 64-6198 was 130- to 3500-fold selective for the OFQ/N receptor (ORL1) compared with opiate receptors. In the cAMP inhibition assay, Ro 64-6198 was a full agonist at the ORL1 and a partial agonist at the mu opiate receptor. When human embryonic kidney 293 cells stably expressing the human ORL1 receptor were pre-exposed (30 min) to either OFQ/N or Ro 64-6198, the ability of both agonists to inhibit forskolin-mediated cAMP accumulation was strongly reduced, indicating a functional desensitization of the second messenger cascade. However, acidic washes of OFQ/N-exposed cells fully restored the sensitivity of the ORL1 receptor for agonists. In contrast, the cAMP response in Ro 64-6198-exposed cells remained impaired after acidic washes, suggesting sustained receptor internalization at 30 min. In agreement with this finding, the number of cell-surface ORL1 receptors was significantly reduced after Ro 64-6198 pre-exposure, and this effect could be blocked with high sucrose concentrations. When Ro 64-6198 was chronically administered to rats, no signs of tolerance to its anxiolytic-like effects were detected following 15 days of daily drug exposure. In agreement with the behavioral results, Ro 64-6198 was able to reduce brain ORL1 binding sites in both acutely and chronically treated rats. Full recovery of ORL1 binding sites was observed 24 h after Ro 64-6198 administration with a t1/2 of approximately 5.5 h. These data show that nonpeptide agonists at the ORL1 receptor have a good clinical potential as anxiolytics without causing tolerance.


Subject(s)
Imidazoles/pharmacology , Opioid Peptides/agonists , Receptors, Opioid/drug effects , Spiro Compounds/pharmacology , Anxiety/psychology , Body Temperature/drug effects , Brain Chemistry/drug effects , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Drug Tolerance , Humans , Kidney/metabolism , Opioid Peptides/genetics , Pain Measurement/drug effects , Radiopharmaceuticals , Receptors, Opioid/genetics , Transfection , Weight Gain/drug effects , Nociceptin Receptor , Nociceptin
7.
Neuropharmacology ; 41(1): 97-107, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11445190

ABSTRACT

Identification of synthetic agonists and antagonists at orphan receptors represents an important step for understanding their physiological function and therapeutic potential. Accordingly, we have recently described a non-peptide agonist at the opioid receptor like (ORL1) receptor (1S,3aS)-8-(2,3,3a,4,5,6-hexahydro-1H-phenalen-1-yl)-1-phenyl-1,3,8-triazaspiro[4.5]decan-4-one (Ro64-6198; Jenck et al., PNAS 94 (2000) 4938; Wichmann et al., Eur. J. Med. Chem. 35 (2000) 839). We have investigated the effects of this compound in various tests of rodent neurological function, utilising ORL1 knockout mice to examine the pharmacological specificity of Ro64-6198. In male C57BL/6J mice, effects on balance and motor co-ordination were detected following low doses (0.3-1mg/kg IP) of Ro64-6198. At higher doses (1-3mg/kg IP), effects on swim behaviour and hypothermia was observed. At 10mg/kg, each effect became more profound and a severe neurological disturbance appeared, including loss of righting reflex. These effects of Ro64-6198 (10mg/kg IP) were absent in ORL1 receptor knockout mice. In male, hooded Lister rats, Ro64-6198 (6-10mg/kg IP), produced some disturbance of neurological function, including hypoactivity, rotarod performance, grip strength and mild hypothermia. An impairment of food responding under a variable interval (VI) 20s schedule of reinforcement was noted at 3mg/kg. These results confirm Ro64-6198 to be a highly selective pharmacological tool to investigate ORL1 receptor function in vivo and, furthermore, that activation of this receptor is accompanied by a variety of effects on neurological function.


Subject(s)
Imidazoles/pharmacology , Neurons/drug effects , Receptors, Opioid/agonists , Spiro Compounds/pharmacology , Animals , Autoradiography , Body Temperature/drug effects , Conditioning, Operant/drug effects , Hand Strength/physiology , Image Processing, Computer-Assisted , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Postural Balance/drug effects , Posture/physiology , Rats , Receptors, Opioid/genetics , Nociceptin Receptor
8.
Naunyn Schmiedebergs Arch Pharmacol ; 363(5): 551-5, 2001 May.
Article in English | MEDLINE | ID: mdl-11383716

ABSTRACT

The pharmacological profile of the non-peptide OP4 receptor (ORL1, LC132) agonist, Ro 64-6198, was investigated, in three electrically stimulated nociceptin/orphanin FQ (NC)-sensitive preparations, namely the mouse and rat vas deferens and the guinea pig ileum. Ro 64-6198 mimicked the inhibitory effect of NC in the three preparations, while showing slow kinetics of action and a slowly reversible effect compared to the fast and immediately and completely reversible effect of the natural peptide. Ro 64-6198 displayed similar pEC50 and Emax values as NC in the mouse and rat vas deferens while it was 100-fold less potent but more efficacious (higher Emax) than NC in the guinea pig ileum. In the rat vas deferens the effects of Ro 64-6198 were antagonised by [Nphe1]NC(1-13)NH2 and J-113397 with pKB values (6.30 and 8.05, respectively) similar to those obtained against NC (6.20 and 7.77, respectively). Naloxone (1 microM) was inactive. In the guinea pig ileum a clear shift of the concentration response curve to Ro 64-6198 was obtained only using a cocktail of antagonists (naloxone + [Nphe1]NC(1-13)NH2 or naloxone + J-113397). In the mouse vas deferens the antagonists were inactive against Ro 64-6198 either when tested alone or in combination. Therefore, Ro 64-6198 behaved as a selective OP4 receptor agonist only in the rat tissue. These results suggest a physiological heterogeneity in OP4 receptors across tissues and species and may explain why, when tested in vivo, Ro 64-6198 mimics the potent anxiolytic effect of NC better in the rat than in the mouse.


Subject(s)
Anti-Anxiety Agents/pharmacology , Imidazoles/pharmacology , Opioid Peptides/pharmacology , Spiro Compounds/pharmacology , Vasodilator Agents/pharmacology , Animals , Dose-Response Relationship, Drug , Electric Stimulation , Guinea Pigs , Ileum/drug effects , Ileum/physiology , Male , Mice , Organ Specificity/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Opioid/agonists , Receptors, Opioid/physiology , Species Specificity , Vas Deferens/drug effects , Vas Deferens/physiology , Nociceptin Receptor , Nociceptin
9.
Braz J Med Biol Res ; 34(2): 145-54, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11175489

ABSTRACT

This article is a transcription of an electronic symposium sponsored by the Brazilian Society of Neuroscience and Behavior (SBNeC). Invited researchers from the European Union, North America and Brazil discussed two issues on anxiety, namely whether panic is a very intense anxiety or something else, and what aspects of clinical anxiety are reproduced by animal models. Concerning the first issue, most participants agreed that generalized anxiety and panic disorder are different on the basis of clinical manifestations, drug response and animal models. Also, underlying brain structures, neurotransmitter modulation and hormonal changes seem to involve important differences. It is also common knowledge that existing animal models generate different types of fear/anxiety. A challenge for future research is to establish a good correlation between animal models and nosological classification.


Subject(s)
Anxiety , Disease Models, Animal , Panic , Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Anxiety/physiopathology , Benzodiazepines/pharmacology , Brain/drug effects , Brain/physiopathology , Fear/drug effects , Humans , Panic/drug effects , Periaqueductal Gray/drug effects , Periaqueductal Gray/physiopathology , Serotonin/pharmacology
10.
Braz. j. med. biol. res ; 34(2): 145-154, Feb. 2001.
Article in English | LILACS | ID: lil-281592

ABSTRACT

This article is a transcription of an electronic symposium sponsored by the Brazilian Society of Neuroscience and Behavior (SBNeC). Invited researchers from the European Union, North America and Brazil discussed two issues on anxiety, namely whether panic is a very intense anxiety or something else, and what aspects of clinical anxiety are reproduced by animal models. Concerning the first issue, most participants agreed that generalized anxiety and panic disorder are different on the basis of clinical manifestations, drug response and animal models. Also, underlying brain structures, neurotransmitter modulation and hormonal changes seem to involve important differences. It is also common knowledge that existing animal models generate different types of fear/anxiety. A challenge for future research is to establish a good correlation between animal models and nosological classification


Subject(s)
Humans , Anxiety , Disease Models, Animal , Panic , Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Anxiety/physiopathology , Benzodiazepines/pharmacology , Brain/drug effects , Brain/physiopathology , Computer Communication Networks , Fear/drug effects , Panic/drug effects , Periaqueductal Gray/drug effects , Periaqueductal Gray/physiopathology , Serotonin/pharmacology
11.
Behav Pharmacol ; 11(3-4): 257-68, 2000 Jun.
Article in English | MEDLINE | ID: mdl-11103880

ABSTRACT

Neonatal ventral hippocampal lesions in the rat result in post-pubertal onset of behavioural abnormalities, modelling some aspects of schizophrenia. We further assessed the behavioural effects of neonatal lesions in rats in a variety of cognitive tasks and in the prepulse inhibition (PPI) of startle response paradigm. Prepubescent, lesioned rats exhibited startle responses and PPI similar to controls whereas, at adulthood, they showed a deficit in PPI. Lesioned rats acquired both passive and active avoidance responses. However, compared to controls, they showed a deficit in passive avoidance retention and in acquisition of active avoidance responses. In a cued Morris water-maze task, lesioned rats demonstrated adequate sensorimotor functions and appropriate motivation to escape from water. However, they were impaired in place learning and in remembering the location of a submerged platform. In conclusion, neonatal ventral hippocampal lesions result in the post-pubertal emergence of long-lasting deficits in sensorimotor gating and in the capacity to acquire and retain information in tests of spatial and avoidance learning. Therefore, this neurodevelopmental model of schizophrenia seems to exhibit an interesting degree of validity in possibly simulating some cognitive impairments and sensorimotor gating deficits frequently observed in psychotic patients.


Subject(s)
Hippocampus/pathology , Learning Disabilities/psychology , Maze Learning , Schizophrenia/physiopathology , Animals , Avoidance Learning , Cognition Disorders/etiology , Disease Models, Animal , Hippocampus/growth & development , Rats , Rats, Sprague-Dawley , Reflex, Startle/physiology
13.
Eur J Med Chem ; 35(9): 839-51, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11006485

ABSTRACT

The development of 8-(2,3,3a,4,5, 6-hexahydro-1H-phenalen1-yl)-1-phenyl-1,3,8-triaza-spiro[4. 5]decan-4-ones 3 starting from (RS)-8-acenaphten-1-yl-1-phenyl-1,3, 8-triazaspiro[4.5]decan-4-one 1 is reported. The synthesis and the binding affinities at human OFQ and opioid (micro, kappa, delta) receptors of the stereoisomers 3a-f are described. In vitro the most selective compound, (1S,3aS)-8-(2,3,3a,4,5, 6-hexahydro-1H-phenalen1-yl)-1-phenyl-1,3,8-triaza-spiro[4. 5]decan-4-one 3c, was found to act as a full agonist at the OFQ receptor in the GTPgamma(35)S binding test. It turned out to be selective versus a variety of other neurotransmitter systems. When tested in vivo following intraperitoneal injection, compound 3c was found to decrease neophobia in a novel environment and to exhibit dose-dependent anxiolytic-like effects in the elevated plus-maze procedure, thus confirming the effects observed following intracerebroventricular infusion of the OFQ peptide in rat.


Subject(s)
Anti-Anxiety Agents/chemical synthesis , Anti-Anxiety Agents/pharmacology , Aza Compounds/chemical synthesis , Aza Compounds/pharmacology , Phenalenes , Receptors, Opioid/agonists , Spiro Compounds/chemical synthesis , Spiro Compounds/pharmacology , Alprazolam/pharmacology , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Injections, Intraperitoneal , Male , Maze Learning/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Opioid/drug effects , Nociceptin Receptor
14.
J Neurosci ; 20(11): 4037-49, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10818139

ABSTRACT

We have used site-directed mutagenesis in conjunction with homologous recombination to generate two mouse lines carrying point mutations in the glycine binding site of the NMDAR1 subunit (Grin1). Glycine concentration-response curves from acutely dissociated hippocampal neurons revealed a 5- and 86-fold reduction in receptor glycine affinity in mice carrying Grin1(D481N) and Grin1(K483Q) mutations, respectively, whereas receptor glutamate affinity remained unaffected. Homozygous mutant Grin1(D481N) animals are viable and fertile and appear to develop normally. However, homozygous mutant Grin1(K483Q) animals are significantly lighter at birth, do not feed, and die within a few days. No gross abnormalities in CNS anatomy were detected in either Grin1(D481N) or Grin1(K483Q) mice. Interestingly, in situ hybridization and Western blot analysis revealed changes in the expression levels of NMDA receptor subunits in Grin1(D481N) mice relative to wild type that may represent a compensatory response to the reduction in receptor glycine affinity. Grin1(D481N) mice exhibited deficits in hippocampal theta burst-induced long-term potentiation (LTP) and spatial learning and also a reduction in sensitivity to NMDA-induced seizures relative to wild-type controls, consistent with a reduced activation of NMDA receptors. Mutant mice exhibited normal prepulse inhibition but showed increased startle reactivity. Preliminary analysis indicated that the mice exhibit a decreased natural aversion to an exposed environment. The lethal phenotype of Grin1(K483Q) animals confirms the critical role of NMDA receptor activation in neonatal survival. A milder reduction in receptor glycine affinity results in an impairment of LTP and spatial learning and alterations in anxiety-related behavior, providing further evidence for the role of NMDA receptor activation in these processes.


Subject(s)
Glycine/physiology , Point Mutation/physiology , Receptors, Glycine/genetics , Receptors, Glycine/physiology , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Autoradiography , Behavior, Animal/physiology , Blotting, Southern , Blotting, Western , Calcium/physiology , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Gene Targeting , Hippocampus/cytology , Hippocampus/metabolism , Homozygote , Image Interpretation, Computer-Assisted , In Situ Hybridization , Long-Term Potentiation/physiology , Mice , Patch-Clamp Techniques , Point Mutation/genetics , Reflex, Startle/physiology , Reverse Transcriptase Polymerase Chain Reaction , Seizures/chemically induced , Seizures/genetics , Seizures/physiopathology
15.
Bioorg Med Chem Lett ; 10(8): 831-4, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10782696

ABSTRACT

We have investigated 8-cycloalkyl-1-phenyl-1,3,8-triaza-spiro[4.5]decan-4-o nes as ligands for the ORL1 receptor. These unsophisticated, achiral compounds show remarkable affinity for the ORL1 receptor. Optimizing for selectivity we show that the maximum of affinity and selectivity versus the other opioid receptors is achieved for 8-cyclodecyl-1-phenyl-1,3,8-triaza-spiro[4.5]decan-4-o ne 2e and 8-(cis-4-isopropyl-cyclohexyl)-1-phenyl-1,3,8-triaza-spiro[4.5] decan-4-one 2q. The identified compounds (2e, 2q) are more or less equipotent to the natural ligand itself, both in the binding assay and in the functional GTPgammaS assay.


Subject(s)
Receptors, Opioid/metabolism , Triazines/chemistry , Triazines/pharmacology , Cell Line , Humans , Ligands , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Structure-Activity Relationship , Nociceptin Receptor
16.
Proc Natl Acad Sci U S A ; 97(9): 4938-43, 2000 Apr 25.
Article in English | MEDLINE | ID: mdl-10758169

ABSTRACT

The biochemical and behavioral effects of a nonpeptidic, selective, and brain-penetrant agonist at the ORL1 receptor are reported herein. This low molecular weight compound [(1S,3aS)-8- (2,3,3a,4,5, 6-hexahydro-1H-phenalen-1-yl)-1-phenyl-1,3,8-triaza- spiro[4. 5]decan-4-one] has high affinity for recombinant human ORL1 receptors and has 100-fold selectivity for ORL1 over other members of the opioid receptor family. It is a full agonist at these receptors and elicits dose-dependent anxiolytic-like effects in a set of validated models of distinct types of anxiety states in the rat (i.e., elevated plus-maze, fear-potentiated startle, and operant conflict). When given systemically, the compound has an efficacy and potency comparable to those of a benzodiazepine anxiolytic such as alprazolam or diazepam. However, this compound is differentiated from a classical benzodiazepine anxiolytic by a lack of efficient anti-panic-like activity, absence of anticonvulsant properties, and lack of effects on motor performance and cognitive function at anxiolytic doses (0.3 to 3 mg/kg i.p.). No significant change in intracranial self-stimulation performance and pain reactivity was observed in this dose range. Higher doses of this compound (>/=10 mg/kg) induced disruption in rat behavior. These data confirm the notable anxiolytic-like effects observed at low doses with the orphanin FQ/nociceptin neuropeptide given locally into the brain and support a role for orphanin FQ/nociceptin in adaptive behavioral fear responses to stress.


Subject(s)
Anti-Anxiety Agents/pharmacology , Fear/physiology , Imidazoles/pharmacology , Maze Learning/drug effects , Receptors, Opioid/agonists , Reflex, Startle/drug effects , Spiro Compounds/pharmacology , Acoustic Stimulation , Alprazolam/pharmacology , Animals , Cognition/drug effects , Cognition/physiology , Conflict, Psychological , Diazepam/pharmacology , Dose-Response Relationship, Drug , Electroshock , Epilepsy/chemically induced , Epilepsy/physiopathology , Fear/drug effects , Humans , Male , Maze Learning/physiology , Pain/physiopathology , Pentylenetetrazole , Rats , Rats, Sprague-Dawley , Receptors, Opioid/physiology , Recombinant Proteins/metabolism , Seizures/chemically induced , Seizures/physiopathology , Self Stimulation/drug effects , Nociceptin Receptor
17.
J Med Chem ; 43(7): 1329-38, 2000 Apr 06.
Article in English | MEDLINE | ID: mdl-10753470

ABSTRACT

The discovery of 8-(5,8-dichloro-1,2,3,4-tetrahydro-naphthalen-2-yl)-1-phenyl-1,3,8-triazaspiro[4.5]decan-4-one, 1a, as a high-affinity ligand for the human ORL1 (orphanin FQ/nociceptin) receptor led to the synthesis of a series of optimized ligands. These compounds exhibit high affinity for the human ORL1 receptor, exhibit moderate to good selectivity versus opioid receptors, and behave as full agonists in biochemical assays. In this paper we present the synthesis, structure-activity relationship (SAR), and biochemical characterization of substituted 1-phenyl-1,3,8-triazaspiro[4.5]decan-4-ones culminating in the discovery of 8-(5-methyl-1,2,3,4-tetrahydro-naphthalen-1-yl)-1-phenyl-1,3,8-triazaspiro[4.5]decan-4-one, 1p, and 8-acenaphten-1-yl-1-phenyl-1,3,8-triazaspiro[4.5]decan-4-one 1q, two high-affinity, potent ORL1 receptor agonists with good to moderate selectivity versus the other opioid receptors.


Subject(s)
Imidazoles/chemical synthesis , Receptors, Opioid/agonists , Spiro Compounds/chemical synthesis , Binding, Competitive , Cell Line , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Ligands , Radioligand Assay , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Structure-Activity Relationship , Nociceptin Receptor
18.
Proc Natl Acad Sci U S A ; 96(18): 10444-9, 1999 Aug 31.
Article in English | MEDLINE | ID: mdl-10468628

ABSTRACT

The neuropeptide orphanin FQ (also known as nociceptin; OFQ/N) has been implicated in modulating stress-related behavior. OFQ/N was demonstrated to reverse stress-induced analgesia and possess anxiolytic-like activity after central administration. To further study physiological functions of OFQ/N, we have generated OFQ/N-deficient mice by targeted disruption of the OFQ/N gene. Homozygous mice display increased anxiety-like behavior when exposed to a novel and threatening environment. OFQ/N-null mice show elevated basal pain threshold but develop normal stress-induced analgesia. Interestingly, these mice show impaired adaptation to repeated stress when compared with wild-type mice, whereas their performance in spatial learning remained unaffected. Basal and poststress plasma corticosterone levels were found to be elevated in OFQ/N-deficient animals. Thus, OFQ/N appears to be crucially involved in the neurobiological regulation of stress-coping behavior and fear.


Subject(s)
Maze Learning , Motor Activity , Opioid Peptides/genetics , Opioid Peptides/physiology , Pain/physiopathology , Stress, Psychological/genetics , Analgesia , Animals , Anxiety/genetics , Anxiety/physiopathology , Corticosterone/blood , Genetic Predisposition to Disease , Heterozygote , Homozygote , Mice , Mice, Knockout , Opioid Peptides/deficiency , Perception , Receptors, Opioid/physiology , Space Perception , Stress, Psychological/physiopathology , Nociceptin Receptor , Nociceptin
19.
Bioorg Med Chem Lett ; 9(16): 2343-8, 1999 Aug 16.
Article in English | MEDLINE | ID: mdl-10476866

ABSTRACT

A series of 8-acenaphthen-1-yl-1-phenyl-1,3,8-triaza-spiro[4.5]decan+ ++-4-one derivatives 1 was studied with respect to the binding affinity for the orphanin FQ (OFQ) and opioid (mu, kappa, delta) receptors. The influence of stereochemistry as well as the substitution pattern of the phenyl-ring in position 1 on the affinity for the orphanin FQ receptor and selectivity to opioid (mu, kappa, delta) receptors is discussed. The most interesting compound 1c was tested for its anxiolytic-like properties in vivo.


Subject(s)
Imidazoles/pharmacology , Receptors, Opioid/agonists , Spiro Compounds/pharmacology , Animals , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/pharmacology , Cell Line , Cricetinae , Exploratory Behavior/drug effects , Humans , Imidazoles/chemistry , Rats , Spiro Compounds/chemistry , Nociceptin Receptor
20.
J Psychopharmacol ; 13(2): 166-70, 1999.
Article in English | MEDLINE | ID: mdl-10475723

ABSTRACT

Acute systemic administration of the selective serotonin (5-HT)1A receptor full agonist flesinoxan enhanced the sensitivity of rats to the panic-like aversion elicited by local stimulation of the dorsolateral periaqueductal grey (dPAG). This experimental paradigm in rats has previously been validated as a simulation of acute anxiety with particular relevance to panic disorder. The dose-dependent decrease in threshold for acute fear responses recorded in rats following intraperitoneal administration of flesinoxan (1-10 mg/kg) was similar to that induced by the panic precipitating agent yohimbine and opposite to the threshold increase induced by the antipanic drug alprazolam. The proaversive effect of flesinoxan observed in rats is consistent with the reported aggravation of the condition of panic patients following oral flesinoxan treatment. Thus, the model adequately detects drug-induced panicogenic-like properties. Data suggest that selective activation of 5-HT1A receptors (pre- and/or post-synaptic in brain and/or periphery) following systemic administration of 5-HT1A receptor full agonists exacerbates aversion in animals or patients with panic anxiety; activation of these receptor subtypes may probably mediate the panicogenic action reported under certain circumstances with non-selective 5-HT mimetics.


Subject(s)
Arousal/drug effects , Avoidance Learning/drug effects , Panic/drug effects , Piperazines/pharmacology , Receptors, Serotonin/drug effects , Serotonin Receptor Agonists/pharmacology , Animals , Dose-Response Relationship, Drug , Electric Stimulation , Male , Periaqueductal Gray/drug effects , Rats , Rats, Wistar , Receptors, Serotonin, 5-HT1
SELECTION OF CITATIONS
SEARCH DETAIL
...