Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Behav Brain Res ; 313: 208-213, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27421830

ABSTRACT

The Wistar-Kyoto (WKY) rat has been proposed as a model of anxiety vulnerability as it exhibits pronounced behavioral inhibition, passive avoidance, exaggerated startle response, enhanced HPA-axis activation, and active avoidance that is resistant to extinction. Accumulating evidence suggests that WKY rats respond differently to rewarding stimuli when compared to outbred strains of rat. Conditioned responding to drug-associated cues is linked with alterations in the activation of mu opioid receptors (MOR) and kappa opioid receptors (KOR) in the nucleus accumbens (NAc). Furthermore, alterations in KOR expression/activation in the NAc of WKY rats are implicated in the regulation of some of the components that make up the unique behavioral phenotype of this strain. The purpose of this study was to extend upon previous work from our laboratory by investigating conditioned morphine reward in adult male WKY and SD rats, and to examine levels of KOR mRNA and MOR mRNA in the NAc at baseline and after acquisition of morphine CPP. Our results demonstrate that SD rats displayed morphine-induced CPP to each of the six doses of morphine tested (0.5, 1.25, 2.5, 5, 7.5, or 10mg/kg). Interestingly, WKY rats demonstrated CPP for only the 1.25, 2.5, and 5mg/kg doses, yet no preference at the lowest (0.5mg/kg) or highest (7.5 and 10mg/kg) doses. qPCR analysis of MOR and KOR in the NAc revealed no strain differences in basal levels of MOR, but higher levels of KOR in WKY rats compared to those of SD rats. Interestingly, after completion of the CPP task, WKY rats had overall higher levels of NAc MOR mRNA compared to SD rats; the initial basal differences in NAc KOR levels persisted without change due to CPP in either strain. These results demonstrate that the WKY rat exhibits a unique pattern of behavioral responding to morphine and implicates differences in NAc KOR signaling as a potential source of aversion to higher doses of morphine. Additionally, the CPP-induced upregulation of NAc MOR mRNA in WKY rats warrants further investigation in terms of its potential role as a factor constituting a unique vulnerability to subsequent drug exposure.


Subject(s)
Cues , Morphine/pharmacology , Nucleus Accumbens/metabolism , RNA, Messenger/metabolism , Receptors, Opioid, mu/metabolism , Animals , Conditioning, Operant/drug effects , Male , Nucleus Accumbens/drug effects , Rats, Inbred WKY , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/genetics
2.
Schizophr Bull ; 42(5): 1270-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27056716

ABSTRACT

Receptors for antipsychotics in the hypothalamus contribute to antipsychotics-induced weight gain; however, many of these receptors are also expressed in the intestine. The role of these intestinally-expressed receptors, and their potential modulation of nutrient absorption, have not been investigated in the context of antipsychotics-induced weight gain. Here we tested the effect of dietary fructose and intestinal fructose uptake on clozapine-induced weight gain in mice. Weight gain was determined in wild type mice and mice lacking the GLUT5 fructose transporter that were "orally-administered" 20mg/kg clozapine for 28 days. To assess the role of dietary fructose, clozapine-treated mice were fed controlled diets with different levels of fructose. Effect of clozapine treatment on intestinal fructose transport activity and expression levels of various receptors that bind clozapine, as well as several genes involved in gluconeogenesis and lipogenesis were measured using real-time RT-PCR and western blotting. Oral administration of clozapine significantly increased body weight in wild type C57BL/6 mice but not in GLUT5 null mice. The clozapine-induced weight gain was proportional to the percentage of fructose in the diet. Clozapine-treated mice increased intestinal fructose uptake without changing the intestinal expression level of GLUT5. Clozapine-treated mice expressed significantly higher levels of intestinal H1 histamine receptor in the wild type but not GLUT5 null mice. Clozapine also increased the intestinal expression of fructokinase and several genes involved in gluconeogenesis and lipogenesis. Our results suggest that increased intestinal absorption and metabolism of fructose contributes to clozapine-induced weight gain. Eliminating dietary fructose might prevent antipsychotics-induced weight gain.


Subject(s)
Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Dietary Carbohydrates/metabolism , Fructose/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Intestinal Absorption/drug effects , Weight Gain/drug effects , Animals , Antipsychotic Agents/administration & dosage , Clozapine/administration & dosage , Disease Models, Animal , Fructokinases/metabolism , Glucose Transporter Type 5 , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
PLoS One ; 10(11): e0143908, 2015.
Article in English | MEDLINE | ID: mdl-26619275

ABSTRACT

Several studies have shown that repeated stressful experiences during childhood increases the likelihood of developing depression- and anxiety-related disorders in adulthood; however, the underlying mechanisms are not well understood. We subjected drd3-EGFP and drd3-null mice to daily, two hour restraint stress episodes over a five day period during preadolescence (postnatal day 35 to 39), followed by social isolation. When these mice reached adulthood (post-natal day > 90), we assessed locomotor behavior in a novel environment, and assessed depression-related behavior in the Porsolt Forced Swim test. We also measured the expression and function of dopamine D3 receptor in limbic brain areas such as hippocampus, nucleus accumbens and amygdala in control and stressed drd3-EGFP mice in adulthood. Adult male mice subjected to restraint stress during preadolescence exhibited both anxiety- and depression-related behaviors; however, adult female mice subjected to preadolescent restraint stress exhibited only depression-related behaviors. The development of preadolescent stress-derived psychiatric disorders was blocked by D3 receptor selective antagonist, SB 277011-A, and absent in D3 receptor null mice. Adult male mice that experienced stress during preadolescence exhibited a loss of D3 receptor expression and function in the amygdala but not in hippocampus or nucleus accumbens. In contrast, adult female mice that experienced preadolescent stress exhibited increased D3 receptor expression in the nucleus accumbens but not in amygdala or hippocampus. Our results suggest that the dopamine D3 receptor is centrally involved in the etiology of adult anxiety- and depression-related behaviors that arise from repeated stressful experiences during childhood.


Subject(s)
Anxiety/genetics , Depression/genetics , Receptors, Dopamine D3/genetics , Receptors, Dopamine D3/metabolism , Stress, Psychological/genetics , Animals , Anxiety/metabolism , Anxiety/physiopathology , Brain/metabolism , Depression/metabolism , Depression/physiopathology , Disease Models, Animal , Female , Male , Mice , Nitriles/pharmacology , Sex Factors , Social Isolation/psychology , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Tetrahydroisoquinolines/pharmacology
4.
Front Psychiatry ; 6: 106, 2015.
Article in English | MEDLINE | ID: mdl-26257661

ABSTRACT

Dysregulation of brain-derived neurotrophic factor (BDNF), behavioral inhibition temperament (BI), and small hippocampal volume have been linked to anxiety disorders. Individuals with BI show facilitated acquisition of the classically conditioned eyeblink response (CCER) as compared to non-BI individuals, and a similar pattern is seen in an animal model of BI, the Wistar-Kyoto (WKY) rat. The present study examined the role of hippocampal BDNF in the facilitated delay CCER of WKY rats. Consistent with earlier work, acquisition was facilitated in WKY rats compared to the Sprague Dawley (SD) rats. Facilitated acquisition was associated with increased BDNF, TrkB, and Arc mRNA in the dentate gyrus of SD rats, but learning-induced increases in BDNF and Arc mRNA were significantly smaller in WKY rats. To determine whether reduced hippocampal BDNF in WKY rats was a contributing factor for their facilitated CCER, BDNF or saline infusions were given bilaterally into the dentate gyrus region 1 h prior to training. BDNF infusion did not alter the acquisition of SD rats, but significantly dampened the acquisition of CCER in the WKY rats, such that acquisition was similar to SD rats. Together, these results suggest that inherent differences in the BDNF system play a critical role in the facilitated associative learning exhibited by WKY rats, and potentially individuals with BI. Facilitated associative learning may represent a vulnerability factor in the development of anxiety disorders.

5.
Pharmacol Res ; 99: 174-84, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26116441

ABSTRACT

The dopamine D3 receptor exhibits agonist-dependent tolerance and slow response termination (SRT) signaling properties that distinguish it from the closely-related D2 receptors. While amino acid residues important for D3 receptor ligand binding have been identified, the residues involved in activation of D3 receptor signaling and induction of signaling properties have not been determined. In this paper, we used cis and trans isomers of a novel D3 receptor agonist, 8-OH-PBZI, and site-directed mutagenesis to identify key residues involved in D3 receptor signaling function. Our results show that trans-8-OH-PBZI, but not cis-8-OH-PBZI, elicit the D3 receptor tolerance and SRT properties. We show that while both agonists require a subset of residues in the orthosteric binding site of D3 receptors for activation of the receptor, the ability of the two isomers to differentially induce tolerance and SRT is mediated by interactions with specific residues in the sixth transmembrane helix and third extracellular loop of the D3 receptor. We also show that unlike cis-8-OH-PBZI, which is a partial agonist at the dopamine D2S receptor and full agonist at dopamine D2L receptor, trans-8-OH-PBZI is a full agonist at both D2S and D2L receptors. The different effect of the two isomers on D3 receptor signaling properties and D2S receptor activation correlated with differential effects of the isomers on agonist-induced mouse locomotor activity. The two isomers of 8-OH-PBZI represent novel pharmacological tools for in silico D3 and D2 receptor homology modeling and for determining the role of D3 receptor tolerance and SRT properties in signaling and behavior.


Subject(s)
Receptors, Dopamine D3/chemistry , Receptors, Dopamine D3/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites/genetics , Cell Line , Dopamine Agonists/chemistry , Dopamine Agonists/pharmacology , Humans , Indoles/chemistry , Indoles/pharmacology , Male , Mice , Mice, Transgenic , Models, Molecular , Molecular Sequence Data , Motor Activity/drug effects , Mutagenesis, Site-Directed , Protein Conformation , Receptors, Dopamine D3/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Stereoisomerism , Tetrahydronaphthalenes/pharmacology
6.
Eur J Neurosci ; 42(2): 1849-57, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25900179

ABSTRACT

The dopamine D1 receptor is centrally involved in mediating the effects of cocaine and is essential for cocaine-induced locomotor sensitization. Changes in D1 receptor expression have been reported in various models of cocaine addiction; however, the mechanisms that mediate these changes in D1 receptor expression are not well understood. Using preadolescent drd1a-EGFP mice and a binge cocaine treatment protocol we demonstrate that the D1 receptor is post-transcriptionally regulated in the caudate-putamen of cocaine-sensitized animal. While cocaine-sensitized mice express high levels of steady-state D1 receptor mRNA, the expression of D1 receptor protein is not elevated. We determined that the post-transcriptional regulation of D1 receptor mRNA is rapidly attenuated and D1 receptor protein levels increase within 30 min when the sensitized mice are challenged with cocaine. The rapid increase in D1 receptor protein levels requires de novo protein synthesis and correlates with the cocaine-induced hyperlocomotor activity in the cocaine-sensitized mice. The increase in D1 receptor protein levels in the caudate-putamen inversely correlated with the levels of microRNA 142-3p and 382, both of which regulate D1 receptor protein expression. The levels of these two microRNAs decreased significantly within 5 min of cocaine challenge in sensitized mice. The results provide novel insights into the previously unknown rapid kinetics of D1 receptor protein expression which occurs in a time scale that is comparable to the expression of immediate early genes. Furthermore, the results suggest a potential novel role for inherently labile microRNAs in regulating the rapid expression of D1 receptor protein in cocaine-sensitized animals.


Subject(s)
Caudate Nucleus/drug effects , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Nucleus Accumbens/drug effects , Receptors, Dopamine D1/metabolism , Animals , Animals, Newborn , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Transgenic , MicroRNAs/metabolism , Motor Activity/drug effects , Motor Activity/genetics , RNA, Messenger/metabolism , Receptors, Dopamine D1/genetics
7.
Neurosci Lett ; 585: 33-7, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25445374

ABSTRACT

Chronic treatment with levodopa or antipsychotics results in manifestation of side-effects such as dyskinesia which correlates with changes in expression and function of receptors and signaling proteins. Previous studies have suggested a role for the dopamine D3 receptor in Parkinson's disease (PD) and tardive dyskinesia. Yet the expression and signaling function of D3 receptor in these disorders is not well understood. Here we tested the hypothesis that chronic levodopa treatment alters both expression and function of D3 receptors in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine plus probenecid (MPTP/p) mouse model of PD. drd3-EGFP reporter mice were injected biweekly with saline or MPTP and probenecid for a 5-week period. During the last two weeks of the 5-week period, the mice were administered saline or levodopa twice daily. Locomotor activity was measured during the treatment period. D3 receptor expression was determined by western blot analysis. D3 receptor signaling function was determined at tissue and single cell level by measuring the activation of D3 receptor-mitogen activated protein kinase (MAPK) pathway. The drd3-EGFP mice administered MPTP/p exhibited akinesia/bradykinesia. Expression of D3 receptor protein in the dorsal striatum specifically increased in the MPTP/p-treated mice administered levodopa. In the dorsal striatum of levodopa and MPTP/p-treated drd3-EGFP mice, administration of a D3 receptor-selective dose of agonist, PD128907, failed to activate D3 receptor-MAPK signaling. These results suggest that MPTP-induced lesion and chronic levodopa treatment alters D3 receptor expression and function in the dorsal striatum which could contribute to the development of dyskinesias and other motor side-effects.


Subject(s)
Antiparkinson Agents/pharmacology , Levodopa/pharmacology , Parkinsonian Disorders/drug therapy , Receptors, Dopamine D3/metabolism , Animals , Antiparkinson Agents/therapeutic use , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Levodopa/therapeutic use , Mice, Mutant Strains , Motor Activity/drug effects , Parkinsonian Disorders/etiology , Parkinsonian Disorders/metabolism , Receptors, Dopamine D3/genetics
8.
Behav Brain Res ; 263: 46-50, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24462727

ABSTRACT

L-DOPA-induced dyskinesias (LID) are motor side effects associated with treatment of Parkinson's disease (PD). The etiology of LID is not clear; however, studies have shown that the dopamine D3 receptor is upregulated in the basal ganglia of mice, rats and non-human primate models of LID. It is not known if the upregulation of D3 receptor is a cause or result of LID. In this paper we tested the hypothesis that overexpression of the dopamine D3 receptor in dorsal striatum, in the absence of dopamine depletion, will elicit LID. Replication-deficient recombinant adeno-associated virus-2 expressing the D3 receptor or enhanced green fluorescent protein (EGFP) were stereotaxically injected, unilaterally, into the dorsal striatum of adult rats. Post-hoc immunohistochemical analysis revealed that ectopic expression of the D3 receptor was limited to neurons near the injection sites in the dorsal striatum. Following a 3-week recovery period, rats were administered saline, 6 mg/kg L-DOPA, 0.1 mg/kg PD128907 or 10 mg/kg ES609, i.p., and motor behaviors scored. Rats overexpressing the D3 receptor specifically exhibited contralateral axial abnormal involuntary movements (AIMs) following administration of L-DOPA and PD128907 but not saline or the novel agonist ES609. Daily injection of 6 mg/kg L-DOPA to the rats overexpressing the D3 receptor also caused increased vacuous chewing behavior. These results suggest that overexpression of the D3 receptor in the dorsal striatum results in the acute expression of agonist-induced axial AIMs and chronic L-DOPA-induced vacuous chewing behavior. Agonists such as ES609 might provide a novel therapeutic approach to treat dyskinesia.


Subject(s)
Corpus Striatum/metabolism , Dyskinesia, Drug-Induced/metabolism , Neurons/metabolism , Receptors, Dopamine D3/metabolism , Animals , Antiparkinson Agents/adverse effects , Benserazide/pharmacology , Benzopyrans/pharmacology , Dependovirus , Dopamine/metabolism , Dopamine Agonists/pharmacology , Female , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Levodopa/adverse effects , Mastication/drug effects , Motor Activity/drug effects , Motor Activity/physiology , Oxazines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D3/genetics , Transfection
9.
Neurosci Lett ; 558: 20-5, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24095672

ABSTRACT

In adult mice, repeated cocaine administration induces behavioral sensitization measured as increased horizontal locomotor activity. Cocaine-induced locomotor sensitization has been well characterized in adult mice. In adult animals, the D1 dopamine receptor is important for mediating effects of cocaine. The effect of cocaine on D1 receptor expression and function in preadolescent animals is less understood. The recently described drd1-enhanced green fluorescent protein (drd1-EGFP) reporter mouse is a useful model for performing such mechanistic studies; however, preadolescent drd1-EGFP mice have not been characterized previously. Here we studied cocaine-induced locomotor sensitization in preadolescent drd1-EGFP reporter mice. We administered 15mg/kg cocaine three times daily at 1h intervals for seven consecutive days beginning on postnatal day 23 to drd1-EGFP reporter mice and the commonly used C57BL/6 mice. Under this regimen, preadolescent mice of both strains exhibited cocaine-induced locomotor sensitization; however, by day 7 the cocaine-induced locomotor activity in the drd1-EGFP mice was maintained for a longer duration compared to the C57BL/6 mice. The preadolescent drd1-EGFP mice also exhibited elevated basal locomotor activity in a novel environment and had higher D1 and D2 dopamine receptor mRNA levels in the caudate nucleus compared to the C57BL/6 mice. The cocaine-induced locomotor sensitization was not retained when the drd1-EGFP mice were maintained cocaine-free for two weeks suggesting that in preadolescent drd1-EGFP mice the cocaine-induced changes do not persist.


Subject(s)
Cocaine/pharmacology , Green Fluorescent Proteins/genetics , Motor Activity/drug effects , Receptors, Dopamine D1/genetics , Age Factors , Animals , Caudate Nucleus/metabolism , Genes, Reporter , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , RNA, Messenger/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Species Specificity , Time Factors
10.
Neuropharmacology ; 79: 359-67, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24316466

ABSTRACT

The D3 dopamine receptor has the highest affinity for dopamine, many antipsychotics as well as agonists used to treat Parkinson's disease and related disorders. We and others have reported that the D3 receptor exhibits a tolerance property wherein repeated agonist stimulation of the receptor results in a progressive loss of agonist-induced signaling response. Recently we reported that the D3 receptor tolerance property is agonist dependent and identified a novel agonist, ES609, which does not elicit D3 receptor tolerance. Here, we used the classical tolerance-inducing D3 receptor agonist, PD128907, and the novel agonist, ES609, to demonstrate that the D3 receptor tolerance property is exhibited not only in cellular signaling in vitro and in vivo, but also manifests at the behavior level. Using AtT-20 cells stably expressing D3 receptors we show that PD128907, but not ES609, induces tolerance in the D3 receptor-mitogen activated protein kinase (MAPK) pathway. Using the novel drd3-EGFP reporter mice, we demonstrate that 0.05 mg/kg PD128907 and 10 mg/kg ES609 selectively activate the D3 receptor-MAPK signaling pathway in vivo; however, only PD128907 induces tolerance. Locomotor behavior assessment showed that both PD128907 and ES609 decreased locomotor activity of the drd3-EGFP mice. While the agonist-induced decrease in locomotor activity was attenuated in drd3-EGFP mice administered two sequential doses of tolerance-inducing agonist PD128907, this attenuation was not seen in mice repeatedly administered the novel agonist, ES609. Together the results suggest that the D3 receptor tolerance property is exhibited in MAPK signaling in vitro and in vivo and also affects agonist-induced locomotor behavior.


Subject(s)
Benzopyrans/pharmacology , Butylamines/pharmacology , Dopamine Agonists/pharmacology , Drug Tolerance , Oxazines/pharmacology , Receptors, Dopamine D3/agonists , Animals , Cell Line, Tumor , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Corpus Striatum/drug effects , Corpus Striatum/physiology , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Phosphorylation/drug effects , Receptors, Dopamine D3/genetics , Receptors, Dopamine D3/metabolism
11.
EMBO Mol Med ; 5(9): 1402-14, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23873704

ABSTRACT

Alcohol addiction is a major social and health concern. Here, we determined the expression profile of microRNAs (miRNAs) in the nucleus accumbens (NAc) of rats treated with alcohol. The results suggest that multiple miRNAs were aberrantly expressed in rat NAc after alcohol injection. Among them, miR-382 was down-regulated in alcohol-treated rats. In both cultured neuronal cells in vitro and in the NAc in vivo, we identified that the dopamine receptor D1 (Drd1) is a direct target gene of miR-382. Via this target gene, miR-382 strongly modulated the expression of DeltaFosB. Moreover, overexpression of miR-382 significantly attenuated alcohol-induced up-regulation of DRD1 and DeltaFosB, decreased voluntary intake of and preference for alcohol and inhibited the DRD1-induced action potential responses. The results indicated that miRNAs are involved in and may represent novel therapeutic targets for alcoholism.


Subject(s)
Alcoholism/genetics , Gene Expression Regulation , MicroRNAs/metabolism , Animals , Ethanol/metabolism , Gene Expression Profiling , MicroRNAs/genetics , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiopathology , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Receptors, Dopamine D1/biosynthesis
12.
ACS Chem Neurosci ; 4(6): 940-51, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23477444

ABSTRACT

The D3 dopamine receptor is a therapeutic target for treating various nervous system disorders such as schizophrenia, Parkinson's disease, depression, and addictive behaviors. The crystal structure of the D3 receptor bound to an antagonist was recently described; however, the structural features that contribute to agonist-induced conformational changes and signaling properties are not well understood. We have previously described the conformation-dependent tolerance and slow response termination (SRT) signaling properties of the D3 receptor and identified the C147 residue in the second intracellular loop (IL2) of the D3 receptor as important for the tolerance property. Interestingly, while IL2 and the C147 residue, in particular, were important for dopamine- and quinpirole-induced tolerance, this residue did not affect the severe tolerance induced by the high affinity, D3 receptor-selective agonist, PD128907. Here, we used D2/D3 receptor chimeras and site-specific D3 receptor mutants to identify another residue, D187, in the second extracellular loop (EC2) of the human D3 receptor that mediates the tolerance property induced by PD128907, quinpirole, pramipexole, and dopamine. Molecular dynamics simulations confirmed the distinct conformation adopted by D3 receptor during tolerance and suggested that in the tolerant D3 receptor the D187 residue in EC2 forms a salt bridge with the H354 residue in EC3. Indeed, site-directed mutation of the H354 residue resulted in loss of PD1287907-induced tolerance. The mapping of specific amino acid residues that contribute to agonist-dependent conformation changes and D3 receptor signaling properties refines the agonist-bound D3 receptor pharmacophore model which will help develop novel D3 receptor agonists.


Subject(s)
Dopamine Agonists/pharmacology , Drug Tolerance/physiology , Extracellular Fluid/physiology , Receptors, Dopamine D3/agonists , Receptors, Dopamine D3/genetics , Amino Acid Sequence , Animals , Benzopyrans/pharmacology , Cells, Cultured , Humans , Mice , Oxazines/pharmacology , Protein Structure, Secondary , Receptors, Dopamine D3/chemistry
13.
Naunyn Schmiedebergs Arch Pharmacol ; 386(2): 97-105, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23160988

ABSTRACT

This study aims to determine the effect of the novel D(3) dopamine receptor agonist, D-264, on activation of D(3) and D(2) dopamine receptor signal transduction pathways and cell proliferation. AtT-20 neuroendocrine cells stably expressing human D(2S), D(2L), and D(3) dopamine receptors were treated with D-264 and the coupling of the receptors to mitogen-activated protein kinase (MAPK) and G protein-coupled inward rectifier potassium (GIRK) channels was determined using Western blotting and whole-cell voltage clamp recording, respectively. D-264 potently activated MAPK signaling pathway coupled to D(2S), D(2L), and D(3) dopamine receptors. The activation of MAPK was more pronounced than the reference agonist quinpirole and was longer lasting. D-264 also activated GIRK channels coupled to D(2S), D(2L), and D(3) receptors. In addition, D-264 dose-dependently induced cell proliferation in AtT-D(2L) and AtT-D(3) cells. These results indicate that D-264 robustly activates GIRK channels and MAPK coupled to D(2) and D(3) dopamine receptors in AtT-20 cells. D-264 is also a potent inducer of cell proliferation.


Subject(s)
Biphenyl Compounds/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Mitogen-Activated Protein Kinases/metabolism , Piperazines/pharmacology , Receptors, Dopamine D2/physiology , Receptors, Dopamine D3/agonists , Thiazoles/pharmacology , Animals , Benzothiazoles , CHO Cells , Cell Line , Cell Proliferation/drug effects , Cloning, Molecular , Cricetinae , Cricetulus , Dopamine Agonists/pharmacology , Humans , Mice
14.
PLoS One ; 7(11): e49288, 2012.
Article in English | MEDLINE | ID: mdl-23152889

ABSTRACT

The D1 dopamine receptor subtype is expressed in the brain, kidney and lymphocytes. D1 receptor function has been extensively studied and the receptor has been shown to modulate a wide range of physiological functions and behaviors. The expression of D1 receptor is known to change during development, disease states and chronic treatment; however, the molecular mechanisms that mediate the changes in D1 receptor expression under these circumstances are not well understood. While previous studies have identified extracellular factors and signaling mechanisms regulating the transcription of D1 receptor gene, very little is known about other regulatory mechanisms that modulate the expression of the D1 receptor gene. Here we report that the D1 receptor is post-transcriptionally regulated during postnatal mouse brain development and in the mouse CAD catecholaminergic neuronal cell line. We demonstrate that this post-transcriptional regulation is mediated by a molecular mechanism involving noncoding RNA. We show that the 1277 bp 3'untranslated region of D1 receptor mRNA is necessary and sufficient for mediating the post-transcriptional regulation. Using deletion and site-directed mutagenesis approaches, we show that the D1 receptor post-transcriptional regulation is specifically mediated by microRNA miR-142-3p interacting with a single consensus binding site in the 1277 bp 3'untranslated region of D1 receptor mRNA. Inhibiting endogenous miR-142-3p in CAD cells increased endogenous D1 receptor protein expression levels. The increase in D1 receptor protein levels was biologically significant as it resulted in enhanced D1 receptor-mediated signaling, determined by measuring the activation of both, adenylate cyclase and, the dopamine- and cAMP-regulated phosphoprotein, DARPP-32. We also show that there is an inverse correlation between miR-142-3p levels and D1 receptor protein expression in the mouse brain during postnatal development. This is the first study to demonstrate that the post-transcriptional regulation of D1 receptor expression is mediated by microRNA-induced translational suppression.


Subject(s)
Gene Expression Regulation , MicroRNAs/metabolism , Receptors, Dopamine D1/genetics , Transcription, Genetic , 3' Untranslated Regions/genetics , Animals , Animals, Newborn , Base Pairing/genetics , Base Sequence , Brain/drug effects , Brain/growth & development , Brain/metabolism , Catecholamines/metabolism , Cell Line , Gene Expression Regulation/drug effects , Genes, Reporter , Male , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Molecular Sequence Data , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Dopamine D1/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic/drug effects
15.
PLoS One ; 7(7): e41623, 2012.
Article in English | MEDLINE | ID: mdl-22911828

ABSTRACT

Soluble cytokine receptors are normal constituents of body fluids that regulate peripheral cytokine and lymphoid activity and whose levels are increased in states of immune activation. Soluble interleukin-6 receptor (sIL-6R) levels positively correlate with disease progression in some autoimmune conditions and psychiatric disorders. Particularly strong links between levels of sIL-6R and the severity of psychotic symptoms occur in schizophrenia, raising the possibility that sIL-6R is involved in this disease. However, there is no evidence that peripheral sIL-6R induces relevant behavioral disturbances. We showed that single subcutaneous injections of sIL-6R (0-1 µg), stimulated novelty stress-induced exploratory motor behaviors in male Balb/c mice within 20-40-min of injection. A progressive increase in vertical stereotypies was observed 40-80 min post injection, persisting for the remainder of the test session. Paralleling these stimulant-like effects, sIL-6R pre-treatment significantly enhanced stereotypy scores following challenge with GBR 12909. We found that peripherally administered sIL-6R crossed the blood-brain barrier, localizing in brain regions associated with cortico-striatal-thalamo-cortical (CSTC) circuits, which are putative neuroanatomical substrates of disorders associated with repetitive stereotypies. Peripherally administered sIL-6R co-localized with gp130, a transmembrane protein involved in IL-6 trans-signaling, in the nucleus accumbens, caudate-putamen, motor and infralimbic cortices, and thalamic nuclei, but not with gp130 in the ventral tegmental area, substantia nigra, or sensorimotor cortex,. The results suggest that peripheral sIL-6R can act as a neuroimmune messenger, crossing the blood brain barrier (BBB) to selectively target CSTC circuits rich in IL-6 trans-signaling protein, and inducing repetitive stereotypies. As such sIL-6R may represent a novel therapeutic agent for relevant psychiatric disorders.


Subject(s)
Cerebral Cortex/metabolism , Cytokine Receptor gp130/metabolism , Motor Activity , Neostriatum/metabolism , Nerve Net/metabolism , Receptors, Interleukin-6/metabolism , Thalamus/metabolism , Animals , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Humans , Injections, Subcutaneous , Male , Mice , Mice, Inbred BALB C , Motor Activity/drug effects , Neostriatum/drug effects , Nerve Net/drug effects , Piperazines/pharmacology , Protein Transport/drug effects , Receptors, Interleukin-6/administration & dosage , Solubility/drug effects , Staining and Labeling , Thalamus/drug effects
16.
Brain Struct Funct ; 217(4): 809-33, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22286951

ABSTRACT

Among dopamine receptors, the expression and function of the D3 receptor subtype is not well understood. The receptor has the highest affinity for dopamine and many drugs that target dopamine receptors.In this paper, we examined, at the single cell level, the characteristics of D3 receptor-expressing cells isolated from different brain regions of male and female mice that were either 35 or 70 days old. The brain regions included nucleus accumbens, Islands of Calleja, olfactory tubercle,retrosplenial cortex, dorsal subiculum, mammillary body,amygdala and septum. The expression analysis was done in the drd3-enhanced green fluorescent protein transgenic mice that report the endogenous expression of D3 receptor mRNA. Using single cell reverse transcriptase PCR, we determined if the D3 receptor-expressing fluorescent cells in these mice were neurons or glia and if they were glutamatergic, GABAergic or catecholaminergic. Next, we determined if the fluorescent cells co-expressed the four other dopamine receptor subtypes, adenylate cyclase V(ACV) isoform, and three different isoforms of G protein coupled inward rectifier potassium (GIRK) channels. The results suggest that D3 receptor is expressed in neurons,with region-specific expression in glutamatergic and GABAergic neurons. The D3 receptor primarily coexpressed with D1 and D2 dopamine receptors with regional, sex and age-dependent differences in the coexpression pattern. The percentage of cells co-expressing D3 receptor and ACV or GIRK channels varied significantly by brain region, sex and age. The molecular characterization of D3 receptor-expressing cells in mouse brain reported here will facilitate the characterization of D(3) receptor function in physiology and pathophysiology.


Subject(s)
Brain/cytology , Brain/metabolism , Neurons/cytology , Neurons/metabolism , Receptors, Dopamine D3/biosynthesis , Animals , Dopamine/metabolism , Female , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Male , Memory/physiology , Mice , Mice, Transgenic , Models, Animal , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Dopamine D3/genetics , Receptors, Dopamine D3/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Single-Cell Analysis/methods , Structure-Activity Relationship
17.
J Biol Chem ; 286(37): 32454-63, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21803776

ABSTRACT

Activation of D1 dopamine receptors expressed in the kidneys promotes the excretion of sodium and regulates sodium levels during increases in dietary sodium intake. A decrease in the expression or function of D1 receptors results in increased sodium retention which can potentially lead to the development of hypertension. Studies have shown that in the absence of functional D1 receptors, in null mice, the systolic, diastolic, and mean arterial pressures are higher. Previous studies have shown that the expression and function of D1 receptors in the kidneys are decreased in animal models of diabetes. The mechanisms that down-regulate the expression of renal D1 receptor gene in diabetes are not well understood. Using primary renal cells and acutely isolated kidneys from the streptozotocin-induced rat diabetic model, we demonstrate that the renal D1 receptor expression is down-regulated by the extracellular cAMP-adenosine pathway in vitro and in vivo. In cultures of primary renal cells, a 3 mm, 60-h cAMP treatment down-regulated the expression of D1 receptors. In vivo, we determined that the plasma and urine cAMP levels as well as the expression of 5'-ectonucleotidase, tissue-nonspecific alkaline phosphatase, and adenosine A2a receptors are significantly increased in diabetic rats. Inhibitors of 5'-ectonucleotidase and tissue-nonspecific alkaline phosphatase, α,ß-methyleneadenosine 5'-diphosphate, and levamisole, respectively, blocked the down-regulation of D1 receptors in the primary renal cells and in the kidney of diabetic animals. The results suggest that inhibitors of the extracellular cAMP-adenosine pathway reverse the down-regulation of renal D1 receptor in diabetes.


Subject(s)
Adenosine/pharmacology , Cyclic AMP/pharmacology , Diabetes Mellitus, Experimental/metabolism , Down-Regulation/drug effects , Kidney/metabolism , Receptors, Dopamine/biosynthesis , Adenosine/metabolism , Alkaline Phosphatase/metabolism , Animals , Antirheumatic Agents/pharmacology , Cyclic AMP/metabolism , Levamisole/pharmacology , Male , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A/metabolism
18.
Biochem Pharmacol ; 79(6): 897-907, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-19879251

ABSTRACT

The D3 but not D2 dopamine receptors exhibit a tolerance property in which agonist-induced D3 receptor response progressively decreases upon repeated agonist stimulation. We have previously shown that the D3 receptor tolerance property is not mediated by receptor internalization, persistent agonist binding or a decrease in receptor binding affinity. In this paper, we test the hypothesis that alterations in D3 receptor conformation underlie the tolerance property. Structural models of wild type and mutant human D3 receptors were generated using the beta adrenergic receptor crystal structure as a template. These models suggested that the agonist-bound D3 receptor undergoes conformational changes that could underlie its tolerance property. To experimentally assess changes in receptor conformation, we measured the accessibility of native cysteine residues present in the extracellular and transmembrane regions of the human D3 receptor to two different thiol-modifying biotinylating reagents. The accessibilities of the native cysteine residues present in the D3 receptor were assessed under control conditions, in the presence of agonist and under conditions that induced receptor tolerance. By comparing the accessibility of D3 receptor cysteine residues to hydrophobic and hydrophilic thiol-modifying biotinylating reagents, we show that the alteration of D3 receptor conformation during tolerance involves the net movement of cysteine residues into a hydrophobic environment. Our results show that the conformation state of the D3 receptor during tolerance is distinct from the conformation under basal and agonist-bound conditions. The results suggest that the D3 receptor tolerance property is mediated by conformational changes that may uncouple the receptor from G-protein signaling.


Subject(s)
Dopamine/pharmacology , Receptors, Dopamine D3/metabolism , Signal Transduction , Animals , Biotinylation , Cell Line , Computer Simulation , Humans , Mice , Models, Biological , Molecular Biology , Mutation , Patch-Clamp Techniques , Protein Conformation , Receptors, Dopamine D3/chemistry , Receptors, Dopamine D3/genetics , Structure-Activity Relationship , Sulfhydryl Compounds/metabolism
19.
J Neurochem ; 110(3): 1058-69, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19493166

ABSTRACT

The generation of dopamine (DA) neurons from stem cells holds great promise in the treatment of Parkinson's disease and other neural disease associated with dysfunction of DA neurons. Mesenchymal stem cells (MSCs) derived from the adult bone marrow show plasticity with regards to generating cells of other germ layers. In addition to reduced ethical concerns, MSCs could be transplanted across allogeneic barriers, making them desirable stem cells for clinical applications. We have reported on the generation of DA cells from human MSCs using sonic hedgehog (SHH), fibroblast growth factor 8 and basic fibroblast growth factor. Despite the secretion of DA, the cells did not show evidence of functional neurons, and were therefore designated DA progenitors. Here, we report on the role of brain-derived neurotrophic factor (BDNF) in the maturation of the MSC-derived DA progenitors. 9-day induced MSCs show significant tropomyosin-receptor-kinase B expression, which correlate with its ligand, BDNF, being able to induce functional maturation. The latter was based on Ca2+ imaging analyses and electrophysiology. BDNF-treated cells showed the following: increases in intracellular Ca2+ upon depolarization and after stimulation with the neurotransmitters acetylcholine and GABA and, post-synaptic currents by electrophysiological analyses. In addition, BDNF induced increased DA release upon depolarization. Taken together, these results demonstrate the crucial role for BDNF in the functional maturation of MSC-derived DA progenitors.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Cell Enlargement , Dopamine/physiology , Mesenchymal Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Adolescent , Adult , Cells, Cultured , Humans , Mesenchymal Stem Cells/cytology , Neurons/cytology , Receptors, Dopamine D1/physiology , Young Adult
20.
Stem Cells ; 25(11): 2797-808, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17656644

ABSTRACT

Dopamine (DA) neurons derived from stem cells are a valuable source for cell replacement therapy in Parkinson disease, to study the molecular mechanisms of DA neuron development, and for screening pharmaceutical compounds that target DA disorders. Compared with other stem cells, MSCs derived from the adult human bone marrow (BM) have significant advantages and greater potential for immediate clinical application. We report the identification of in vitro conditions for inducing adult human MSCs into DA cells. Using a cocktail that includes sonic hedgehog and fibroblast growth factors, human BM-derived MSCs were induced in vitro to become DA cells in 12 days. Based on tyrosine hydroxylase (TH) expression, the efficiency of induction was determined to be approximately 67%. The cells develop a neuronal morphology expressing the neuronal markers NeuN and beta III tubulin, but not glial markers, glial fibrillary acidic protein and Olig2. As the cells acquire a postmitotic neuronal fate, they downregulate cell cycle activator proteins cyclin B, cyclin-dependent kinase 2, and proliferating cell nuclear antigen. Molecular characterization revealed the expression of DA-specific genes such as TH, Pitx3, Nurr1, DA transporter, and vesicular monoamine transporter 2. The induced MSCs also synthesize and secrete DA in a depolarization-independent manner. The latter observation is consistent with the low expression of voltage gated Na(+) and Ca(2+) channels in the induced MSCs and suggests that the cells are at an immature stage of development likely representing DA neuronal progenitors. Taken together, the results demonstrate the ability of adult human BM-derived MSCs to form DA cells in vitro.


Subject(s)
Dopamine/physiology , Mesenchymal Stem Cells/physiology , Phenotype , Adolescent , Adult , Cell Differentiation/genetics , Cells, Cultured , Dopamine/genetics , Flow Cytometry/methods , Genetic Markers/physiology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , RNA, Messenger/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...