Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters










Publication year range
1.
Neuroscience ; 248: 479-87, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23811070

ABSTRACT

D2 receptor null mutant (Drd2(-/-)) mice have altered responses to the rewarding and locomotor effects of psychostimulant drugs, which is evidence of a necessary role for D2 receptors in these behaviors. Furthermore, work with mice that constitutively express only the D2 receptor short form (D2S), as a result of genetic deletion of the long form (D2L), provides the basis for a current model in which D2L is thought to be the postsynaptic D2 receptor on medium spiny neurons in the basal forebrain, and D2S the autoreceptor that regulates the activity of dopamine neurons and dopamine synthesis and release. Because constitutive genetic deletion of the D2 or D2L receptor may cause compensatory changes that influence functional outcomes, our approach is to identify aspects of the abnormal phenotype of a Drd2(-/-) mouse that can be normalized by virus-mediated D2 receptor expression. Drd2(-/-) mice are deficient in basal and methamphetamine-induced locomotor activation and lack D2 receptor agonist-induced activation of G protein-regulated inward rectifying potassium channels (GIRKs) in dopaminergic neurons. Here we show that virus-mediated expression of D2L in the nucleus accumbens significantly restored methamphetamine-induced locomotor activation, but not basal locomotor activity, compared to mice receiving the control virus. It also restored the effect of methamphetamine to decrease time spent in the center of the activity chamber in female but not male Drd2(-/-) mice. Furthermore, the effect of expression of D2S was indistinguishable from D2L. Similarly, virus-mediated expression of either D2S or D2L in substantia nigra neurons restored D2 agonist-induced activation of GIRKs. In this acute expression system, the alternatively spliced forms of the D2 receptor appear to be equally capable of acting as postsynaptic receptors and autoreceptors.


Subject(s)
Locomotion/drug effects , Neurons/metabolism , Nucleus Accumbens/cytology , Receptors, Dopamine D2/metabolism , Animals , Behavior, Animal/drug effects , Dopamine Agonists/pharmacology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Gene Expression Regulation/drug effects , Gene Transfer Techniques , Male , Methamphetamine/pharmacology , Mice , Mice, Knockout , Neurons/drug effects , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/deficiency
2.
J Psychopharmacol ; 24(6): 897-904, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19282420

ABSTRACT

Methylphenidate (MP) and amphetamine (AMPH) are the most frequently prescribed medications for the treatment of attention-deficit/hyperactivity disorder (ADHD). Both drugs are believed to derive their therapeutic benefit by virtue of their dopamine (DA)-enhancing effects, yet an explanation for the observation that some patients with ADHD respond well to one medication but not to the other remains elusive. The dopaminergic effects of MP and AMPH are also thought to underlie their reinforcing properties and ultimately their abuse. Polymorphisms in the human gene that codes for the DA D4 receptor (D4R) have been repeatedly associated with ADHD and may correlate with the therapeutic as well as the reinforcing effects of responses to these psychostimulant medications. Conditioned place preference (CPP) for MP, AMPH and cocaine were evaluated in wild-type (WT) mice and their genetically engineered littermates, congenic on the C57Bl/6J background, that completely lack D4Rs (knockout or KO). In addition, the locomotor activity in these mice during the conditioning phase of CPP was tested in the CPP chambers. D4 receptor KO and WT mice showed CPP and increased locomotor activity in response to each of the three psychostimulants tested. D4R differentially modulates the CPP responses to MP, AMPH and cocaine. While the D4R genotype affected CPP responses to MP (high dose only) and AMPH (low dose only) it had no effects on cocaine. Inasmuch as CPP is considered an indicator of sensitivity to reinforcing responses to drugs these data suggest a significant but limited role of D4Rs in modulating conditioning responses to MP and AMPH. In the locomotor test, D4 receptor KO mice displayed attenuated increases in AMPH-induced locomotor activity whereas responses to cocaine and MP did not differ. These results suggest distinct mechanisms for D4 receptor modulation of the reinforcing (perhaps via attenuating dopaminergic signalling) and locomotor properties of these stimulant drugs. Thus, individuals with D4 receptor polymorphisms might show enhanced reinforcing responses to MP and AMPH and attenuated locomotor response to AMPH.


Subject(s)
Amphetamine/pharmacology , Cocaine/pharmacology , Conditioning, Psychological/drug effects , Methylphenidate/pharmacology , Motor Activity/drug effects , Receptors, Dopamine D4/genetics , Analysis of Variance , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Conditioning, Psychological/physiology , Dopamine Uptake Inhibitors/pharmacology , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Genotype , Male , Mice , Mice, Knockout , Motor Activity/genetics , Random Allocation
3.
Pharmacol Biochem Behav ; 90(3): 387-93, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18456309

ABSTRACT

Alleles of the human dopamine D(4) receptor (D(4)R) gene (DRD4.7) have repeatedly been found to correlate with novelty seeking, substance abuse, pathological gambling, and attention-deficit hyperactivity disorder (ADHD). If these various psychopathologies are a result of attenuated D(4)R-mediated signaling, mice lacking D(4)Rs (D(4)KO) should be more impulsive than wild-type (WT) mice and exhibit more novelty seeking. However, in our study, D(4)KO and WT mice showed similar levels of impulsivity as measured by delay discounting performance and response inhibition on a Go/No-go test, suggesting that D(4)R-mediated signaling may not affect impulsivity. D(4)KO mice were more active than WT mice in the first 5 min of a novel open field test, suggesting greater novelty seeking. For both genotypes, more impulsive mice habituated less in the novel open field. These data suggest that the absence of D(4)Rs is not sufficient to cause psychopathologies associated with heightened impulsivity and novelty seeking.


Subject(s)
Exploratory Behavior/physiology , Impulsive Behavior/genetics , Impulsive Behavior/psychology , Receptors, Dopamine D4/deficiency , Animals , Cues , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Polymorphism, Genetic , Psychomotor Performance/physiology , Receptors, Dopamine D4/genetics
4.
J Comp Physiol B ; 178(2): 167-77, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17912534

ABSTRACT

3-Iodothyronamine is considered as a derivate of thyroid hormone as a result of enzymatic deiodination and decarboxylation. The physiological role of thyronamine (T1AM) is not known. The aim of this study was to analyze the metabolic response to T1AM in the Djungarian hamster Phodopus sungorus. We measured the influence of T1AM (50 mg/kg) on metabolic rate (VO(2)), body temperature (T (b)) and respiratory quotient (RQ) in this species and in BL/6 mice. T1AM treated hamsters as well as the mice showed a rapid decrease in VO(2) and T (b), accompanied by a reduction of RQ from normal values of about approximately 0.9 to approximately 0.70 for several hours. This indicates that carbohydrate utilisation is blocked by the injection of T1AM and that metabolic pathways are rerouted from carbohydrate to lipid utilisation in response to T1AM. This assumption was further supported by the observation that the treatment of T1AM caused ketonuria and a significant loss of body fat. Our results indicate that T1AM has the potential to control the balance between glucose and lipid utilisation in vivo.


Subject(s)
Blood Glucose/metabolism , Lipid Metabolism/physiology , Mice, Inbred C57BL/metabolism , Phodopus/metabolism , Thyronines/metabolism , Animals , Basal Metabolism/drug effects , Basal Metabolism/physiology , Body Composition/drug effects , Body Composition/physiology , Body Temperature/drug effects , Body Temperature/physiology , Cricetinae , Dietary Carbohydrates/metabolism , Dietary Fats/metabolism , Energy Metabolism/drug effects , Energy Metabolism/physiology , Female , Ketones/urine , Male , Mice , Photoperiod , Seasons , Species Specificity , Thyronines/pharmacology
5.
J Pharmacol Exp Ther ; 321(1): 178-86, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17218486

ABSTRACT

The synthetic amines methamphetamine (METH), amphetamine (AMPH), and their metabolite para-hydroxyamphetamine (POHA) are chemically and structurally related to the catecholamine neurotransmitters and a small group of endogenous biogenic amines collectively referred to as the trace amines (TAs). Recently, it was reported that METH, AMPH, POHA, and the TAs para-tyramine (TYR) and beta-phenylethylamine (PEA) stimulate cAMP production in human embryonic kidney (HEK)-293 cells expressing rat trace amine-associated receptor 1 (rTAAR1). The discovery that METH and AMPH activate the rTAAR1 motivated us to study the effect of these drugs on the mouse TAAR1 (mTAAR1) and a human-rat chimera (hrChTAAR1). Furthermore, because S-(+)-isomers of METH and AMPH are reported to be more potent and efficacious in vivo than R-(-), we determined the enantiomeric selectivity of all three species of TAAR1. In response to METH, AMPH, or POHA exposure, the accumulation of cAMP by HEK-293 cells stably expressing different species of TAAR1 was concentration- and isomer-dependent. EC50 values for S-(+)-METH were 0.89, 0.92, and 4.44 microM for rTAAR1, mTAAR1, and h-rChTAAR1, respectively. PEA was a potent and full agonist at each species of TAAR1, whereas TYR was a full agonist for the rodent TAAR1s but was a partial agonist at h-rChTAAR1. Interestingly, both isomers of METH were full agonists at mTAAR1 and h-rChTAAR1, whereas both were partial agonists at rTAAR1. Taken together, these in vitro results suggest that, in vivo, TAAR1 could be a novel mediator of the effects of these drugs.


Subject(s)
Amphetamine/pharmacology , Central Nervous System Stimulants/pharmacology , Methamphetamine/pharmacology , Receptors, G-Protein-Coupled/drug effects , p-Hydroxyamphetamine/pharmacology , Animals , Cell Line , Chimera , Cloning, Molecular , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Mice , Organ Culture Techniques , Plasmids , Rats , Receptors, G-Protein-Coupled/chemistry , Species Specificity , Stereoisomerism
6.
Br J Pharmacol ; 149(8): 967-78, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17088868

ABSTRACT

Classical biogenic amines (adrenaline, noradrenaline, dopamine, serotonin and histamine) interact with specific families of G protein-coupled receptors (GPCRs). The term 'trace amines' is used when referring to p-tyramine, beta-phenylethylamine, tryptamine and octopamine, compounds that are present in mammalian tissues at very low (nanomolar) concentrations. The pharmacological effects of trace amines are usually attributed to their interference with the aminergic pathways, but in 2001 a new gene was identified, that codes for a GPCR responding to p-tyramine and beta-phenylethylamine but not to classical biogenic amines. Several closely related genes were subsequently identified and designated as the trace amine-associated receptors (TAARs). Pharmacological investigations in vitro show that many TAAR subtypes may not respond to p-tyramine, beta-phenylethylamine, tryptamine or octopamine, suggesting the existence of additional endogenous ligands. A novel endogenous thyroid hormone derivative, 3-iodothyronamine, has been found to interact with TAAR1 and possibly other TAAR subtypes. In vivo, micromolar concentrations of 3-iodothyronamine determine functional effects which are opposite to those produced on a longer time scale by thyroid hormones, including reduction in body temperature and decrease in cardiac contractility. Expression of all TAAR subtypes except TAAR1 has been reported in mouse olfactory epithelium, and several volatile amines were shown to interact with specific TAAR subtypes. In addition, there is evidence that TAAR1 is targeted by amphetamines and other psychotropic agents, while genetic linkage studies show a significant association between the TAAR gene family locus and susceptibility to schizophrenia or bipolar affective disorder.


Subject(s)
Receptors, Biogenic Amine/metabolism , Amino Acids/metabolism , Animals , Humans , Ligands , Receptors, G-Protein-Coupled/metabolism
7.
Psychopharmacology (Berl) ; 182(1): 33-44, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16136297

ABSTRACT

RATIONALE: The rewarding effects of lateral hypothalamic brain stimulation, various natural rewards, and several drugs of abuse are attenuated by D1 or D2 dopamine receptor (D1R or D2R) antagonists. Much of the evidence for dopaminergic involvement in rewards is based on pharmacological agents with limited or "relative" selectivity for dopamine receptor subtypes. Genetically engineered animal models provide a complementary approach to pharmacological investigations. OBJECTIVES: In the present study, we explored the contribution of dopamine D2Rs to (1) brain stimulation reward (BSR) and (2) the potentiation of this behavior by morphine and amphetamine using D2R-deficient mice. METHODS: Wild-type (D2Rwt), heterozygous (D2Rhet), and D2R knockout (D2Rko) mice were trained to turn a wheel for rewarding brain stimulation. Once equivalent rate-frequency curves were established, morphine-induced (0, 1.0, 3.0, and 5.6 mg/kg s.c.) and amphetamine-induced (0, 1.0, 2.0, and 4.0 mg/kg i.p.) potentiations of BSR were determined. RESULTS: The D2Rko mice required approximately 50% more stimulation than the D2Rwt mice did. With the equi-rewarding levels of stimulation current, amphetamine potentiated BSR equally across the three genotypes. In contrast, morphine potentiated rewarding stimulation in the D2Rwt, had no effect in the D2Rhet, and antagonized rewarding stimulation in the D2Rko mice. CONCLUSIONS: D2R elimination decreases, but does not eliminate, the rewarding effects of lateral hypothalamic stimulation. After compensation for this deficit, amphetamine continues to potentiate BSR, while morphine does not.


Subject(s)
Brain/drug effects , Hypothalamic Area, Lateral/drug effects , Morphine/pharmacology , Receptors, Dopamine D1/deficiency , Receptors, Dopamine D2/deficiency , Reward , Self Stimulation/drug effects , Amphetamine/pharmacology , Animals , Brain Mapping , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Heterozygote , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motivation , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/genetics
8.
Mol Psychiatry ; 9(7): 718-26, 2004 Jul.
Article in English | MEDLINE | ID: mdl-14699433

ABSTRACT

The dopamine D4 receptor (D4R) is a candidate gene for attention deficit/hyperactivity disorder (ADHD) based on genetic studies reporting that particular polymorphisms are present at a higher frequency in affected children. However, the direct participation of the D4R in the onset or progression of ADHD has not been tested. Here, we generated a mouse model with high face value to screen candidate genes for the clinical disorder by neonatal disruption of central dopaminergic pathways with 6-hydroxydopamine (6-OHDA). The lesioned mice exhibited hyperactivity that waned after puberty, paradoxical hypolocomotor responses to amphetamine and methylphenidate, poor behavioral inhibition in approach/avoidance conflict tests and deficits in continuously performed motor coordination tasks. To determine whether the D4R plays a role in these behavioral phenotypes, we performed 6-OHDA lesions in neonatal mice lacking D4Rs (Drd4(-/-)). Although striatal dopamine contents and tyrosine hydroxylase-positive midbrain neurons were reduced to the same extent in both genotypes, Drd4(-/-) mice lesioned with 6-OHDA did not develop hyperactivity. Similarly, the D4R antagonist PNU-101387G prevented hyperactivity in wild-type 6-OHDA-lesioned mice. Furthermore, wild-type mice lesioned with 6-OHDA showed an absence of behavioral inhibition when tested in the open field or the elevated plus maze, while their Drd4(-/-) siblings exhibited normal avoidance for the unprotected areas of these mazes. Together, our results from a combination of genetic and pharmacological approaches demonstrate that D4R signaling is essential for the expression of juvenile hyperactivity and impaired behavioral inhibition, relevant features present in this ADHD-like mouse model.


Subject(s)
Attention Deficit Disorder with Hyperactivity/genetics , Attention Deficit Disorder with Hyperactivity/physiopathology , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/physiology , Amphetamine/pharmacology , Animals , Animals, Outbred Strains , Attention Deficit Disorder with Hyperactivity/drug therapy , Behavior, Animal/physiology , Central Nervous System Stimulants/pharmacology , Corpus Striatum/cytology , Corpus Striatum/physiopathology , Denervation , Disease Models, Animal , Male , Methylphenidate/pharmacology , Mice , Mice, Knockout , Motor Activity/physiology , Neural Pathways , Oxidopamine , Phenotype , Receptors, Dopamine D4 , Substantia Nigra/cytology , Substantia Nigra/physiopathology , Sympatholytics
9.
Mol Pharmacol ; 60(6): 1181-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11723224

ABSTRACT

The trace amine para-tyramine is structurally and functionally related to the amphetamines and the biogenic amine neurotransmitters. It is currently thought that the biological activities elicited by trace amines such as p-tyramine and the psychostimulant amphetamines are manifestations of their ability to inhibit the clearance of extracellular transmitter and/or stimulate the efflux of transmitter from intracellular stores. Here we report the discovery and pharmacological characterization of a rat G protein-coupled receptor that stimulates the production of cAMP when exposed to the trace amines p-tyramine, beta-phenethylamine, tryptamine, and octopamine. An extensive pharmacological survey revealed that psychostimulant and hallucinogenic amphetamines, numerous ergoline derivatives, adrenergic ligands, and 3-methylated metabolites of the catecholamine neurotransmitters are also good agonists at the rat trace amine receptor 1 (rTAR1). These results suggest that the trace amines and catecholamine metabolites may serve as the endogenous ligands of a novel intercellular signaling system found widely throughout the vertebrate brain and periphery. Furthermore, the discovery that amphetamines, including 3,4-methylenedioxymethamphetamine (MDMA; "ecstasy"), are potent rTAR1 agonists suggests that the effects of these widely used drugs may be mediated in part by this receptor as well as their previously characterized targets, the neurotransmitter transporter proteins.


Subject(s)
Amphetamine/pharmacology , Lysergic Acid Diethylamide/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Receptors, Biogenic Amine/agonists , Amino Acid Sequence , Animals , Catecholamines/metabolism , Catecholamines/pharmacology , Chromosome Mapping , Chromosomes, Human, Pair 6 , Cloning, Molecular , Dopamine Agents/pharmacology , Humans , Molecular Sequence Data , Neurotransmitter Agents/pharmacology , Rats , Receptors, Biogenic Amine/metabolism , Sequence Homology, Amino Acid , Serotonin Agents/pharmacology , Subcellular Fractions , Tumor Cells, Cultured
10.
Hypertension ; 38(3): 303-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566895

ABSTRACT

Polymorphism of the dopamine receptor type-2 (D(2)) gene is associated with essential hypertension. To assess whether D(2) receptors participate in regulation of blood pressure (BP), we studied mice in which the D(2) receptor was disrupted. In anesthetized mice, systolic and diastolic BPs (in millimeters of mercury) were higher in D(2) homozygous and heterozygous mutant mice than in D(2)+/+ littermates. BP after alpha-adrenergic blockade decreased to a greater extent in D(2)-/- mice than in D(2)+/+ mice. Epinephrine excretion was greater in D(2)-/- mice than in D(2)+/+ mice, and acute adrenalectomy decreased BP to a similar level in D(2)-/- and D(2)+/+ mice. An endothelin B (ET[B]) receptor blocker for both ET(B1) and ET(B2) receptors decreased, whereas a selective ET(B1) blocker increased, BP in D(2)-/- mice but not D(2)+/+ mice. ET(B) receptor expression was greater in D(2)-/- mice than in D(2)+/+ mice. In contrast, blockade of ET(A) and V(1) vasopressin receptors had no effect on BP in either D(2)-/- or D(2)+/+ mice. The hypotensive effect of an AT(1) antagonist was also similar in D(2)-/- and D(2)+/+ mice. Basal Na(+),K(+)-ATPase activities in renal cortex and medulla were higher in D(2)+/+ mice than in D(2)-/- mice. Urine flow and sodium excretion were higher in D(2)-/- mice than in D(2)+/+ mice before and after acute saline loading. Thus, complete loss of the D(2) receptor results in hypertension that is not due to impairment of sodium excretion. Instead, enhanced vascular reactivity in the D(2) mutant mice may be caused by increased sympathetic and ET(B) receptor activities.


Subject(s)
Hypertension/physiopathology , Receptors, Adrenergic/physiology , Receptors, Dopamine D2/genetics , Receptors, Endothelin/physiology , Adrenergic alpha-Antagonists/pharmacology , Angiotensin Receptor Antagonists , Animals , Antidiuretic Hormone Receptor Antagonists , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Body Weight , Catechols/urine , Endothelin Receptor Antagonists , Endothelin-1/pharmacology , Female , Genotype , Hypertension/drug therapy , Hypertension/genetics , Losartan/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Oligopeptides/pharmacology , Phentolamine/pharmacology , Piperidines/pharmacology , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptor, Endothelin A , Receptor, Endothelin B , Receptors, Adrenergic/drug effects , Receptors, Dopamine D2/physiology , Receptors, Endothelin/agonists , Sodium/urine , Sodium-Potassium-Exchanging ATPase/metabolism , Urodynamics , Viper Venoms/pharmacology
11.
Neuropsychopharmacology ; 25(4): 527-36, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11557166

ABSTRACT

Phenotypes were assessed topographically in mice lacking functional D(2) dopamine receptors ['knockouts'], using an ethologically based approach to assess all behaviours in the natural repertoire. D(2)-null mice evidenced an ethogram characterised initially by modest reductions in locomotion and shifts in rearing topographies. Subsequently, topographies of behaviour habituated similarly for wildtypes and 'knockouts'. Following challenge with the D(2)-like agonist RU 24213, both inhibition of rearing at a lower dose and induction of stereotyped sniffing and ponderous locomotion at higher doses were essentially absent in D(2)-null mice. Following challenge with the D(1)-like agonist A 68930, vacuous chewing was released in D(2)-null mice. This topographical approach to phenotypic characterisation implicates: (i) the D(2) receptor in these D(2)-like agonist effects and in oppositional D(1)-like: D(2)-like interactions; and (ii) the operation of material compensatory processes consequent to the developmental absence of D(2) receptors which are able to maintain ethological function under tonic, 'naturalistic' conditions but not under 'phasic' challenge.


Subject(s)
Behavior, Animal/physiology , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/physiology , Animals , Chromans/pharmacology , Dopamine Agonists/pharmacology , Female , Grooming/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Phenethylamines/pharmacology , Phenotype
12.
J Neurosci ; 21(16): 5864-70, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11487609

ABSTRACT

The bioactivity of neuropeptides can be regulated by a variety of post-translational modifications, including proteolytic processing. Here, gene-targeted mice producing defective prohormone convertase 2 (PC2) were used to examine the post-translational processing of two neuroendocrine prohormones, pro-opiomelanocortin (POMC) and pro-orphanin FQ (pOFQ)/nociceptin (N), in the brain. Reversed-phase HPLC and gel-exclusion chromatography were combined with specific radioimmunoassays to analyze the processing patterns of these two prohormones in the hypothalamus and the amygdala. In the case of POMC, the lack of PC2 activity completely prevented carboxy-shortening of beta-endorphins and greatly diminished conversion of beta-lipotropin to gamma-lipotropin and beta-endorphin. Although conversion of beta-lipotropin to beta-endorphin decreased, the lack of PC2 activity caused an increase in beta-lipotropin and beta-endorphin levels in the mutant animals, but no increases in POMC or biosynthetic intermediates were seen. The extent of OFQ/N production was significantly lower in PC2-deficient mice and there was an accumulation of relatively large amounts of pOFQ/N and biosynthetic intermediates. These results demonstrate that PC2 is directly involved in the biogenesis of two brain neuropeptides in vivo and suggest that the specific prohormone and cellular context influences neuropeptide processing by PCs.


Subject(s)
Brain/metabolism , Pro-Opiomelanocortin/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational , Receptors, Opioid/metabolism , Subtilisins/biosynthesis , Amygdala/chemistry , Amygdala/metabolism , Animals , Brain Chemistry , Chromatography, Gel , Chromatography, High Pressure Liquid , Gene Targeting , Heterozygote , Homozygote , Hypothalamus/chemistry , Hypothalamus/metabolism , Mice , Mice, Knockout , Proprotein Convertase 2 , Radioimmunoassay , Subtilisins/genetics , beta-Endorphin/biosynthesis , beta-Lipotropin/biosynthesis , beta-Lipotropin/metabolism
13.
J Neurosci ; 21(11): 3756-63, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11356863

ABSTRACT

The dopamine D(4) receptor (D(4)R) is predominantly expressed in the frontal cortex (FC), a brain region that receives dense input from midbrain dopamine (DA) neurons and is associated with cognitive and emotional processes. However, the physiological significance of this dopamine receptor subtype has been difficult to explore because of the slow development of D(4)R agonists and antagonists the selectivity and efficacy of which have been rigorously demonstrated in vivo. We have attempted to overcome this limitation by taking a multidimensional approach to the characterization of mice completely deficient in this receptor subtype. Electrophysiological current and voltage-clamp recordings were performed in cortical pyramidal neurons from wild-type and D(4)R-deficient mice. The frequency of spontaneous synaptic activity and the frequency and duration of paroxysmal discharges induced by epileptogenic agents were increased in mutant mice. Enhanced synaptic activity was also observed in brain slices of wild-type mice incubated in the presence of the selective D(4)R antagonist PNU-101387G. Consistent with greater electrophysiological activity, nerve terminal glutamate density associated with asymmetrical synaptic contacts within layer VI of the motor cortex was reduced in mutant neurons. Taken together, these results suggest that the D(4)R can function as an inhibitory modulator of glutamate activity in the FC.


Subject(s)
Cerebral Cortex/physiopathology , Receptors, Dopamine D2/deficiency , Seizures/physiopathology , 4-Aminopyridine/pharmacology , Animals , Bicuculline/pharmacology , Cerebral Cortex/drug effects , Convulsants/pharmacology , Dopamine/metabolism , Dopamine D2 Receptor Antagonists , Dose-Response Relationship, Drug , Glutamic Acid/metabolism , Immunohistochemistry , In Vitro Techniques , Membrane Potentials/drug effects , Mice , Mice, Neurologic Mutants , Motor Cortex/drug effects , Motor Cortex/metabolism , Motor Cortex/physiopathology , Neural Inhibition/drug effects , Neural Inhibition/genetics , Patch-Clamp Techniques , Piperazines/pharmacology , Presynaptic Terminals/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D4 , Seizures/chemically induced , Sulfonamides/pharmacology
14.
Eur J Neurosci ; 13(5): 995-1001, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11264672

ABSTRACT

According to the dual systems model for opiate reward, dopamine mediates opiate motivation when an animal is in a deprived motivational state (i.e. opiate-dependent and in withdrawal) and not when an animal is in a nondeprived state (i.e. previously drug-naive). To determine the role of the D2 dopamine receptor subtype in mediating opiate motivation, we examined the behaviour of N5 congenic D2 receptor knockout mice and their wild-type siblings in opiate-naive and opiate-dependent and withdrawn place conditioning paradigms. Opiate-naive D2 receptor knockout mice demonstrated acquisition of morphine-conditioned place preference but failed to acquire place preference when conditioned in the deprived state. We propose that D2 receptor function is critical in mediating the motivational effects of opiates only when the animal is in an opiate-dependent and withdrawn motivational state. These findings also underscore the important influence of the genetic background to a given phenotype, as evidenced by the observation that increasing the allelic contribution from the 129/SvJ strain abolishes morphine place preference in C57BL/6 wild-type mice.


Subject(s)
Brain/metabolism , Motivation , Narcotics/pharmacology , Opioid-Related Disorders/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Reward , Substance Withdrawal Syndrome/metabolism , Animals , Conditioning, Psychological/physiology , Female , Male , Mice , Mice, Inbred C57BL/metabolism , Mice, Knockout/metabolism , Opioid-Related Disorders/physiopathology , Substance Withdrawal Syndrome/physiopathology
15.
Proc Natl Acad Sci U S A ; 98(3): 1255-60, 2001 Jan 30.
Article in English | MEDLINE | ID: mdl-11158626

ABSTRACT

The neurotransmitters dopamine (DA) and glutamate in the striatum play key roles in movement and cognition, and they are implicated in disorders of the basal ganglia such as Parkinson's disease. Excitatory synapses in striatum undergo a form of developmental plasticity characterized by a decrease in glutamate release probability. Here we demonstrate that this form of synaptic plasticity is DA and DA D2 receptor dependent. Analysis of spontaneous synaptic responses indicates that a presynaptic mechanism involving inhibition of neurotransmitter release underlies the developmental plasticity. We suggest that a major role of DA in the striatum is to initiate mechanisms that regulate the efficacy of excitatory striatal synapses, producing a decrease in glutamate release.


Subject(s)
Corpus Striatum/physiology , Dopamine/physiology , Excitatory Postsynaptic Potentials/physiology , Neuronal Plasticity/physiology , Receptors, Dopamine D2/physiology , Synapses/physiology , Animals , Corpus Striatum/growth & development , Crosses, Genetic , Excitatory Postsynaptic Potentials/drug effects , Female , Glutamic Acid/physiology , Hydroxydopamines , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/deficiency , Receptors, Dopamine D2/genetics , Species Specificity , Tetrodotoxin/pharmacology , Tyrosine 3-Monooxygenase/metabolism
16.
Proc Natl Acad Sci U S A ; 98(4): 1964-9, 2001 Feb 13.
Article in English | MEDLINE | ID: mdl-11172059

ABSTRACT

Typical neuroleptic drugs elicit their antipsychotic effects mainly by acting as antagonists at dopamine D2 receptors. Much of this activity is thought to occur in the cerebral cortex, where D2 receptors are found largely in inhibitory GABAergic neurons. Here we confirm this localization at the electron microscopic level, but additionally show that a subset of cortical interneurons with low or undetectable expression of D2 receptor isoforms are surrounded by astrocytic processes that strongly express D2 receptors. Ligand binding of isolated astrocyte preparations indicate that cortical astroglia account for approximately one-third of the total D2 receptor binding sites in the cortex, a proportion that we found conserved among rodent, monkey, and human tissues. Further, we show that the D2 receptor-specific agonist, quinpirole, can induce Ca(2+) elevation in isolated cortical astrocytes in a pharmacologically reversible manner, thus implicating this receptor in the signaling mechanisms by which astrocytes communicate with each other as well as with neurons. The discovery of D2 receptors in astrocytes with a selective anatomical relationship to interneurons represents a neuron/glia substrate for cortical dopamine action in the adult cerebral cortex and a previously unrecognized site of action for antipsychotic drugs with affinities at the D2 receptor.


Subject(s)
Astrocytes/metabolism , Prefrontal Cortex/metabolism , Receptors, Dopamine D2/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Binding Sites , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Calcium/metabolism , Cells, Cultured , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Haplorhini , Humans , Ligands , Mice , Neurons/metabolism , Prefrontal Cortex/pathology , Prefrontal Cortex/ultrastructure , Quinpirole/pharmacology , Raclopride/pharmacology , Rats
17.
Proc Natl Acad Sci U S A ; 98(4): 1970-5, 2001 Feb 13.
Article in English | MEDLINE | ID: mdl-11172060

ABSTRACT

The A(2A)R is largely coexpressed with D(2)Rs and enkephalin mRNA in the striatum where it modulates dopaminergic activity. Activation of the A(2A)R antagonizes D(2)R-mediated behavioral and neurochemical effects in the basal ganglia through a mechanism that may involve direct A(2A)R-D(2)R interaction. However, whether the D(2)R is required for the A(2A)R to exert its neural function is an open question. In this study, we examined the role of D(2)Rs in A(2A)R-induced behavioral and cellular responses, by using genetic knockout (KO) models (mice deficient in A(2A)Rs or D(2)Rs or both). Behavioral analysis shows that the A(2A)R agonist 2-4-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamidoadenosine reduced spontaneous as well as amphetamine-induced locomotion in both D(2) KO and wild-type mice. Conversely, the nonselective adenosine antagonist caffeine and the A(2A)R antagonist 8-(3-chlorostyryl)caffeine produced motor stimulation in mice lacking the D(2)R, although the stimulation was significantly attenuated. At the cellular level, A(2A)R inactivation counteracted the increase in enkephalin expression in striatopallidal neurons caused by D(2)R deficiency. Consistent with the D(2) KO phenotype, A(2A)R inactivation partially reversed both acute D(2)R antagonist (haloperidol)-induced catalepsy and chronic haloperidol-induced enkephalin mRNA expression. Together, these results demonstrate that A(2A)Rs elicit behavioral and cellular responses despite either the genetic deficiency or pharmacological blockade of D(2)Rs. Thus, A(2A)R-mediated neural functions are partially independent of D(2)Rs. Moreover, endogenous adenosine acting at striatal A(2A)Rs may be most accurately viewed as a facilitative modulator of striatal neuronal activity rather than simply as an inhibitory modulator of D(2)R neurotransmission.


Subject(s)
Adenosine/analogs & derivatives , Motor Activity/physiology , Receptors, Dopamine D2/physiology , Receptors, Purinergic P1/physiology , Adenosine/pharmacology , Amphetamines/pharmacology , Animals , Caffeine/analogs & derivatives , Caffeine/pharmacology , Catalepsy/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopamine Antagonists/pharmacology , Enkephalins/biosynthesis , Enkephalins/genetics , Gene Expression , Haloperidol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenethylamines/pharmacology , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , RNA, Messenger , Receptor, Adenosine A2A , Receptors, Dopamine D1/biosynthesis , Receptors, Dopamine D2/biosynthesis , Receptors, Purinergic P1/biosynthesis
18.
J Neurophysiol ; 85(2): 659-70, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160501

ABSTRACT

Dopamine (DA) receptors play an important role in the modulation of excitability and the responsiveness of neurons to activation of excitatory amino acid receptors in the striatum. In the present study, we utilized mice with genetic deletion of D2 or D4 DA receptors and their wild-type (WT) controls to examine if the absence of either receptor subtype affects striatal excitatory synaptic activity. Immunocytochemical analysis verified the absence of D2 or D4 protein expression in the striatum of receptor-deficient mutant animals. Sharp electrode current- and whole cell patch voltage-clamp recordings were obtained from slices of receptor-deficient and WT mice. Basic membrane properties were similar in D2 and D4 receptor-deficient mutants and their respective WT controls. In current-clamp recordings in WT animals, very little low-amplitude spontaneous synaptic activity was observed. The frequency of these spontaneous events was increased slightly in D2 receptor-deficient mice. In addition, large-amplitude depolarizations were observed in a subset of neurons from only the D2 receptor-deficient mutants. Bath application of the K+ channel blocker 4-aminopyridine (100 microM) and bicuculline methiodide (10 microM, to block synaptic activity due to activation of GABA(A) receptors) markedly increased spontaneous synaptic activity in receptor-deficient mutants and WTs. Under these conditions, D2 receptor-deficient mice displayed significantly more excitatory synaptic activity than their WT controls, while there was no difference between D4 receptor-deficient mice and their controls. In voltage-clamp recordings, there was an increase in frequency of spontaneous glutamate receptor-mediated inward currents without a change in mean amplitude in D2 receptor-deficient mutants. In WT mice, activation of D2 family receptors with quinpirole decreased spontaneous excitatory events and conversely sulpiride, a D2 receptor antagonist, increased activity. In D2 receptor-deficient mice, sulpiride had very little net effect. Morphologically, a subpopulation of medium-sized spiny neurons from D2 receptor-deficient mice displayed decreased dendritic spines compared with cells from WT mice. These results provide evidence that D2 receptors play an important role in the regulation of glutamate receptor-mediated activity in the corticostriatal or thalamostriatal pathway. These receptors may function as gatekeepers of glutamate release or of its subsequent effects and thus may protect striatal neurons from excessive excitation.


Subject(s)
Corpus Striatum/physiology , Glutamic Acid/physiology , Receptors, Dopamine D2/physiology , Synaptic Transmission/physiology , 4-Aminopyridine/pharmacology , Animals , Corpus Striatum/cytology , Dopamine/pharmacology , Electrophysiology , Immunohistochemistry , In Vitro Techniques , Membranes/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Neurons/drug effects , Neurons/ultrastructure , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/deficiency , Receptors, Dopamine D2/genetics , Synapses/drug effects , Synapses/physiology
19.
Cell Calcium ; 29(1): 39-48, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11133354

ABSTRACT

We analyzed intracellular Ca(2+)and cAMP levels in Chinese hamster ovary cells expressing a cloned rat kappa opioid receptor (CHO-kappa cells). Although expression of kappa(kappa)-opioid receptors was confirmed with a fluorescent dynorphin analog in almost all CHO-kappa cells, the kappa-specific agonists, U50488H or U69593, induced a Ca(2+) transient only in 35% of the cells. The Ca(2+) response occurred in all-or-none fashion and the half-maximal dosage of U50488H (812.1nM) was higher than that (3.2nM) to inhibit forskolin-stimulated cAMP. The kappa-receptors coupled to G(i/o)proteins since pertussis toxin significantly reduced the U50488H actions on intracellular Ca(2+) and cAMP. The Ca(2+) transient originates from IP(3)-sensitive internal stores since the Ca(2+) response was blocked by a PLC inhibitor (U73122) or by thapsigargin depletion of internal stores while removal of extracellular Ca(2+) had no effect. Interestingly, application of dibutyryl cAMP (+ 56.2%) or 8-bromo-cAMP (+ 174.7%) significantly increased the occurrence of U50488H-induced Ca(2+) mobilization while protein kinase A (PKA) inhibitors, Rp-cAMP (-32.3%) or myr-psi PKA (-73.9%) significantly reduced the response. Therefore, it was concluded that cAMP and PKA activity can regulate the Ca(2+) mobilization. These results suggest that the kappa receptor-linked cAMP cascade regulates the occurrence of kappa-opioid-mediated Ca(2+) mobilization.


Subject(s)
Benzeneacetamides , Calcium Signaling/physiology , Calcium/metabolism , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Receptors, Opioid, kappa/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Analgesics/pharmacology , Analgesics, Non-Narcotic/pharmacology , Analgesics, Opioid/pharmacology , Animals , Bucladesine/pharmacology , CHO Cells , Calcium Signaling/drug effects , Cloning, Molecular , Colforsin/pharmacology , Cricetinae , Cyclic AMP/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression/physiology , Peptides , Pyrrolidines/pharmacology , Rats , Receptors, Opioid, kappa/analysis , Receptors, Opioid, kappa/genetics , Thionucleotides/pharmacology , Transfection
20.
Curr Protoc Neurosci ; Chapter 4: Unit 4.2, 2001 May.
Article in English | MEDLINE | ID: mdl-18428481

ABSTRACT

Of the many proteins that are known to be involved in neuronal signaling, one family of gene products, collectively referred to as the G protein-coupled receptors (GPCRs), has received considerable attention. Within the transmembrane domains of GPCRs are clusters of amino acids that tend to be conserved among receptors that bind related ligands. Polymerase chain reaction (PCR)-based approaches to cloning novel GPCRs typically begin with the identification of these well-conserved amino acid motifs, which are then back-translated into degenerate oligonucleotide primers. These pools of degenerate oligonucleotides are the most important variables in PCR cloning of GPCRs. Although GPCRs are used as the focus of this unit, the strategies and techniques described are applicable to the cloning of a wide variety of neuronal gene products. In the first procedure in this unit, either total or poly(A)(+) purified RNA is reverse transcribed into first-strand cDNA. In subsequent steps the cDNA product serves as the template for synthesis and amplification of target receptor sequences by PCR primed with degenerate oligodeoxynucleotides. The product is ready to be cloned and screened as described. Guidelines for database searching are provided to help identify the cloned gene from the known sequence. Typically, only a portion of the receptor coding region is cloned by the above approach. Rapid amplification of cDNA ends (RACE) or anchored PCR is described in this unit and is used to obtain a full-length cDNA amenable for expression studies.


Subject(s)
Cloning, Molecular/methods , Neurons/physiology , Polymerase Chain Reaction/methods , Receptors, G-Protein-Coupled/genetics , Animals , Clone Cells , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Humans , Neurons/metabolism , Receptors, G-Protein-Coupled/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...