Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci ; 36(11): 3106-14, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26985023

ABSTRACT

Repeated exposure to psychostimulants induces locomotor sensitization and leads to persistent changes in the circuitry of the mesocorticolimbic dopamine (DA) system. G-protein-gated inwardly rectifying potassium (GIRK; also known as Kir3) channels mediate a slow IPSC and control the excitability of DA neurons. Repeated 5 d exposure to psychostimulants decreases the size of the GABAB receptor (GABABR)-activated GIRK currents (IBaclofen) in ventral tegmental area (VTA) DA neurons of mice, but the mechanism underlying this plasticity is poorly understood. Here, we show that methamphetamine-dependent attenuation of GABABR-GIRK currents in VTA DA neurons required activation of both D1R-like and D2R-like receptors. The methamphetamine-dependent decrease in GABABR-GIRK currents in VTA DA neurons did not depend on a mechanism of dephosphorylation of the GABAB R2 subunit found previously for other neurons in the reward pathway. Rather, the presence of the GIRK3 subunit appeared critical for the methamphetamine-dependent decrease of GABABR-GIRK current in VTA DA neurons. Together, these results highlight different regulatory mechanisms in the learning-evoked changes that occur in the VTA with repeated exposure to psychostimulants. SIGNIFICANCE STATEMENT: Exposure to addictive drugs such as psychostimulants produces persistent adaptations in inhibitory circuits within the mesolimbic dopamine system, suggesting that addictive behaviors are encoded by changes in the reward neural circuitry. One form of neuroadaptation that occurs with repeated exposure to psychostimulants is a decrease in slow inhibition, mediated by a GABAB receptor and a potassium channel. Here, we examine the subcellular mechanism that links psychostimulant exposure with changes in slow inhibition and reveal that one type of potassium channel subunit is important for mediating the effect of repeated psychostimulant exposure. Dissecting out the components of drug-dependent plasticity and uncovering novel protein targets in the reward circuit may lead to the development of new therapeutics for treating addiction.


Subject(s)
Dopamine Agents/pharmacology , Dopaminergic Neurons/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Methamphetamine/pharmacology , Receptors, GABA-B/metabolism , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/cytology , Animals , Animals, Newborn , Baclofen/pharmacology , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , GABA-B Receptor Agonists/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Vitro Techniques , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, GABA-B/genetics , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/genetics
2.
Proc Natl Acad Sci U S A ; 112(22): 7091-6, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-25964320

ABSTRACT

G protein-gated inwardly rectifying potassium (GIRK) channels are critical regulators of neuronal excitability and can be directly activated by ethanol. Constitutive deletion of the GIRK3 subunit has minimal phenotypic consequences, except in response to drugs of abuse. Here we investigated how the GIRK3 subunit contributes to the cellular and behavioral effects of ethanol, as well as to voluntary ethanol consumption. We found that constitutive deletion of GIRK3 in knockout (KO) mice selectively increased ethanol binge-like drinking, without affecting ethanol metabolism, sensitivity to ethanol intoxication, or continuous-access drinking. Virally mediated expression of GIRK3 in the ventral tegmental area (VTA) reversed the phenotype of GIRK3 KO mice and further decreased the intake of their wild-type counterparts. In addition, GIRK3 KO mice showed a blunted response of the mesolimbic dopaminergic (DA) pathway to ethanol, as assessed by ethanol-induced excitation of VTA neurons and DA release in the nucleus accumbens. These findings support the notion that the subunit composition of VTA GIRK channels is a critical determinant of DA neuron sensitivity to drugs of abuse. Furthermore, our study reveals the behavioral impact of this cellular effect, whereby the level of GIRK3 expression in the VTA tunes ethanol intake under binge-type conditions: the more GIRK3, the less ethanol drinking.


Subject(s)
Dopaminergic Neurons/metabolism , Ethanol/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Ion Channel Gating/physiology , Motivation/genetics , Analysis of Variance , Animals , Binge Drinking/genetics , DNA Primers/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/deficiency , In Situ Hybridization , Ion Channel Gating/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdialysis , Reverse Transcriptase Polymerase Chain Reaction , Reward
3.
J Neurosci ; 35(18): 7131-42, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25948263

ABSTRACT

G-protein-gated inwardly rectifying K(+) (GIRK/Kir3) channel activation underlies key physiological effects of opioids, including analgesia and dependence. GIRK channel activation has also been implicated in the opioid-induced inhibition of midbrain GABA neurons and consequent disinhibition of dopamine (DA) neurons in the ventral tegmental area (VTA). Drug-induced disinhibition of VTA DA neurons has been linked to reward-related behaviors and underlies opioid-induced motor activation. Here, we demonstrate that mouse VTA GABA neurons express a GIRK channel formed by GIRK1 and GIRK2 subunits. Nevertheless, neither constitutive genetic ablation of Girk1 or Girk2, nor the selective ablation of GIRK channels in GABA neurons, diminished morphine-induced motor activity in mice. Moreover, direct activation of GIRK channels in midbrain GABA neurons did not enhance motor activity. In contrast, genetic manipulations that selectively enhanced or suppressed GIRK channel function in midbrain DA neurons correlated with decreased and increased sensitivity, respectively, to the motor-stimulatory effect of systemic morphine. Collectively, these data support the contention that the unique GIRK channel subtype in VTA DA neurons, the GIRK2/GIRK3 heteromer, regulates the sensitivity of the mouse mesolimbic DA system to drugs with addictive potential.


Subject(s)
Analgesics, Opioid/pharmacology , Dopaminergic Neurons/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , GABAergic Neurons/physiology , Motor Activity/physiology , Animals , Dopaminergic Neurons/drug effects , Dose-Response Relationship, Drug , GABAergic Neurons/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Protein Subunits/physiology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology
4.
Proc Natl Acad Sci U S A ; 112(11): 3523-8, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25733865

ABSTRACT

The K(+)/Cl(-) cotransporter (KCC2) allows adult neurons to maintain low intracellular Cl(-) levels, which are a prerequisite for efficient synaptic inhibition upon activation of γ-aminobutyric acid receptors. Deficits in KCC2 activity are implicated in epileptogenesis, but how increased neuronal activity leads to transporter inactivation is ill defined. In vitro, the activity of KCC2 is potentiated via phosphorylation of serine 940 (S940). Here we have examined the role this putative regulatory process plays in determining KCC2 activity during status epilepticus (SE) using knockin mice in which S940 is mutated to an alanine (S940A). In wild-type mice, SE induced by kainate resulted in dephosphorylation of S940 and KCC2 internalization. S940A homozygotes were viable and exhibited comparable basal levels of KCC2 expression and activity relative to WT mice. However, exposure of S940A mice to kainate induced lethality within 30 min of kainate injection and subsequent entrance into SE. We assessed the effect of the S940A mutation in cultured hippocampal neurons to explore the mechanisms underlying this phenotype. Under basal conditions, the mutation had no effect on neuronal Cl(-) extrusion. However, a selective deficit in KCC2 activity was seen in S940A neurons upon transient exposure to glutamate. Significantly, whereas the effects of glutamate on KCC2 function could be ameliorated in WT neurons with agents that enhance S940 phosphorylation, this positive modulation was lost in S940A neurons. Collectively our results suggest that phosphorylation of S940 plays a critical role in potentiating KCC2 activity to limit the development of SE.


Subject(s)
Status Epilepticus/metabolism , Status Epilepticus/pathology , Symporters/metabolism , Animals , Chlorides/metabolism , Endocytosis , Gene Knock-In Techniques , Glutamates/pharmacology , Mice , Mice, Neurologic Mutants , Mutant Proteins/metabolism , Mutation/genetics , Phosphorylation , Phosphoserine/metabolism , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/metabolism , Symporters/genetics , gamma-Aminobutyric Acid/metabolism , K Cl- Cotransporters
5.
Neuron ; 82(3): 659-69, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24811384

ABSTRACT

The subcellular pathways that regulate G protein-gated inwardly rectifying potassium (GIRK or Kir3) channels are important for controlling the excitability of neurons. Sorting nexin 27 (SNX27) is a PDZ-containing protein known to bind GIRK2c/GIRK3 channels, but its function in vivo is poorly understood. Here, we investigated the role of SNX27 in regulating GIRK currents in dopamine (DA) neurons of the ventral tegmental area (VTA). Mice lacking SNX27 in DA neurons exhibited reduced GABABR-activated GIRK currents but had normal Ih currents and DA D2R-activated GIRK currents. Expression of GIRK2a, an SNX27-insensitive splice variant, restored GABABR-activated GIRK currents in SNX27-deficient DA neurons. Remarkably, mice with significantly reduced GABABR-activated GIRK currents in only DA neurons were hypersensitive to cocaine and could be restored to a normal locomotor response with GIRK2a expression. These results identify a pathway for regulating excitability of VTA DA neurons, highlighting SNX27 as a promising target for treating addiction.


Subject(s)
Cocaine/toxicity , G Protein-Coupled Inwardly-Rectifying Potassium Channels/biosynthesis , Sorting Nexins/physiology , Animals , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/antagonists & inhibitors , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Organ Culture Techniques , Protein Binding/genetics , Signal Transduction/genetics , Sorting Nexins/deficiency , Ventral Tegmental Area/cytology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
6.
J Neurosci ; 34(15): 5107-14, 2014 Apr 09.
Article in English | MEDLINE | ID: mdl-24719090

ABSTRACT

G-protein-coupled inwardly rectifying potassium (GIRK) channels contribute to the resting membrane potential of many neurons, including dopamine (DA) neurons in the ventral tegmental area (VTA). VTA DA neurons are bistable, firing in two modes: one characterized by bursts of action potentials, the other by tonic firing at a lower frequency. Here we provide evidence that these firing modes drive bidirectional plasticity of GIRK channel-mediated currents. In acute midbrain slices of mice, we observed that in vitro burst activation of VTA DA neurons potentiated GIRK currents whereas tonic firing depressed these currents. This plasticity was not specific to the metabotropic receptor activating the GIRK channels, as direct activation of GIRK channels by nonhydrolyzable GTP also potentiated the currents. The plasticity of GIRK currents required NMDA receptor and CaMKII activation, and involved protein trafficking through specific PDZ domains of GIRK2c and GIRK3 subunit isoforms. Prolonged tonic firing may thus enhance the probability to switch into burst-firing mode, which then potentiates GIRK currents and favors the return to baseline. In conclusion, activity-dependent GIRK channel plasticity may represent a slow destabilization process favoring the switch between the two firing modes of VTA DA neurons.


Subject(s)
Action Potentials , Dopaminergic Neurons/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Neuronal Plasticity , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dopaminergic Neurons/metabolism , Female , Guanosine Triphosphate/metabolism , Inhibitory Postsynaptic Potentials , Male , Mice , Mice, Inbred C57BL , Protein Isoforms/metabolism , Protein Transport , Receptors, N-Methyl-D-Aspartate/metabolism , Ventral Tegmental Area/cytology , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/physiology
7.
Neuron ; 73(5): 978-89, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22405207

ABSTRACT

Psychostimulants induce neuroadaptations in excitatory and fast inhibitory transmission in the ventral tegmental area (VTA). Mechanisms underlying drug-evoked synaptic plasticity of slow inhibitory transmission mediated by GABA(B) receptors and G protein-gated inwardly rectifying potassium (GIRK/Kir(3)) channels, however, are poorly understood. Here, we show that 1 day after methamphetamine (METH) or cocaine exposure both synaptically evoked and baclofen-activated GABA(B)R-GIRK currents were significantly depressed in VTA GABA neurons and remained depressed for 7 days. Presynaptic inhibition mediated by GABA(B)Rs on GABA terminals was also weakened. Quantitative immunoelectron microscopy revealed internalization of GABA(B1) and GIRK2, which occurred coincident with dephosphorylation of serine 783 (S783) in GABA(B2), a site implicated in regulating GABA(B)R surface expression. Inhibition of protein phosphatases recovered GABA(B)R-GIRK currents in VTA GABA neurons of METH-injected mice. This psychostimulant-evoked impairment in GABA(B)R signaling removes an intrinsic brake on GABA neuron spiking, which may augment GABA transmission in the mesocorticolimbic system.


Subject(s)
Central Nervous System Stimulants/pharmacology , Down-Regulation/drug effects , Methamphetamine/pharmacology , Neurons/drug effects , Receptors, GABA-A/metabolism , Ventral Tegmental Area/cytology , gamma-Aminobutyric Acid/metabolism , Action Potentials/drug effects , Action Potentials/genetics , Animals , Animals, Newborn , Baclofen/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Channelrhodopsins , Dopamine/pharmacology , Dopamine Agents/pharmacology , Drug Interactions , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/ultrastructure , GABA-B Receptor Agonists/pharmacology , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Immunoelectron/methods , Neurons/metabolism , Neurons/ultrastructure , Organophosphorus Compounds/pharmacology , Phosphorylation , Receptors, GABA-A/ultrastructure , Transcription Factors/genetics , Ventral Tegmental Area/drug effects , gamma-Aminobutyric Acid/pharmacology
8.
Psychopharmacology (Berl) ; 203(4): 703-11, 2009 May.
Article in English | MEDLINE | ID: mdl-19050854

ABSTRACT

RATIONALE: We previously colocalized a quantitative trait locus (QTL) for sensitivity to the locomotor stimulant effects of methamphetamine (MA) with a QTL for expression of casein kinase 1 epsilon (Csnk1-epsilon) in the nucleus accumbens (NAc). Subsequently, we identified a single nucleotide polymorphism in CSNK1E (rs135745) that was associated with increased sensitivity to the subjective effects of d-amphetamine in healthy human subjects. Based on these results, we hypothesized that differential expression of Csnk1-epsilon causes differential sensitivity to MA-induced locomotor activity in mice. OBJECTIVE: In the present study, we used PF-670462 (PF), which is a selective inhibitor of Csnk1-epsilon, to directly evaluate the role of Csnk1-epsilon in the locomotor stimulant response to MA in male C57BL/6J mice. METHODS: We administered vehicle, PF, MA, or MA + PF, either via intraperitoneal injections or bilateral intra-NAc microinjections. We also examined Darpp-32 phosphorylation in mice receiving intraperitoneal injections. RESULTS: Intraperitoneal PF (20-40 mg/kg) attenuated the locomotor stimulant response to MA (2 mg/kg) without affecting baseline activity. The high dose of PF also significantly inhibited MA-induced phosphorylation of Darpp-32, providing a potential mechanism by which Csnk1-epsilon contributes to MA-induced locomotor activity. Furthermore, microinjection of PF (5 microg/side) into the NAc completely blocked the locomotor stimulant response to MA (2.5 microg/side) without affecting baseline activity. CONCLUSIONS: These results provide direct evidence that Csnk1-epsilon is crucial for the locomotor stimulant response to a moderate dose of MA and suggest that genetic polymorphisms affecting Csnk1-epsilon expression or function could influence sensitivity to amphetamines in both mice and humans.


Subject(s)
Casein Kinase 1 epsilon/genetics , Central Nervous System Stimulants/pharmacology , Methamphetamine/pharmacology , Motor Activity/drug effects , Animals , Casein Kinase 1 epsilon/antagonists & inhibitors , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Phosphorylation , Polymorphism, Single Nucleotide , Pyrimidines/pharmacology , Quantitative Trait Loci
9.
Behav Genet ; 38(3): 277-91, 2008 May.
Article in English | MEDLINE | ID: mdl-18363093

ABSTRACT

We used short-term selection to produce outbred mouse lines with differences in contextual fear conditioning. Within two generations of selection all low selected mice were homozygous for the recessive tyrc allele and showed the corresponding albino coat color. Freezing differed in the high and low selected lines across a range of parameters. We identified several QTLs for the selection response, including a highly significant QTL at the tyr locus (p < 9.6(-10)). To determine whether the tyrc allele was directly responsible for the response to selection, we examined B6 mice that have a mutant tyr allele (tyr(c-2j-)) and an AJ congenic strain that has the wild-type B6 allele for tyr. These studies showed that the tyr allele had a small influence on fear learning. We used Affymetrix microarrays to identify many differentially expressed genes in the amygdala and hippocampus of the selected lines. We conclude that tyr is one of many alleles that influence fear conditioning.


Subject(s)
Fear , Gene Expression Profiling , Quantitative Trait Loci , Selection, Genetic , Alleles , Amygdala/metabolism , Animals , Conditioning, Psychological , Genes, Recessive , Hippocampus/metabolism , Homozygote , Mice , Mice, Inbred C57BL , Models, Genetic , Oligonucleotide Array Sequence Analysis , Quantitative Trait, Heritable
SELECTION OF CITATIONS
SEARCH DETAIL
...