Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Neurosci ; 39(29): 5634-5646, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31092585

ABSTRACT

Addictive behaviors, including relapse, are thought to depend in part on long-lasting drug-induced adaptations in dendritic spine signaling and morphology in the nucleus accumbens (NAc). While the influence of activity-dependent actin remodeling in these phenomena has been studied extensively, the role of microtubules and associated proteins remains poorly understood. We report that pharmacological inhibition of microtubule polymerization in the NAc inhibited locomotor sensitization to cocaine and contextual reward learning. We then investigated the roles of microtubule end-binding protein 3 (EB3) and SRC kinase in the neuronal and behavioral responses to volitionally administered cocaine. In synaptoneurosomal fractions from the NAc of self-administering male rats, the phosphorylation of SRC at an activating site was induced after 1 d of withdrawal, while EB3 levels were increased only after 30 d of withdrawal. Blocking SRC phosphorylation during early withdrawal by virally overexpressing SRCIN1, a negative regulator of SRC activity known to interact with EB3, abolished the incubation of cocaine craving in both male and female rats. Conversely, mimicking the EB3 increase observed after prolonged withdrawal increased the motivation to consume cocaine in male rats. In mice, the overexpression of either EB3 or SRCIN1 increased dendritic spine density and altered the spine morphology of NAc medium spiny neurons. Finally, a cocaine challenge after prolonged withdrawal recapitulated most of the synaptic protein expression profiles observed at early withdrawal. These findings suggest that microtubule-associated signaling proteins such as EB3 cooperate with actin remodeling pathways, notably SRC kinase activity, to establish and maintain long-lasting cellular and behavioral alterations following cocaine self-administration.SIGNIFICANCE STATEMENT Drug-induced morphological restructuring of dendritic spines of nucleus accumbens neurons is thought to be one of the cellular substrates of long-lasting drug-associated memories. The molecular basis of these persistent changes has remained incompletely understood. Here we implicate for the first time microtubule function in this process, together with key players such as microtubule-bound protein EB3 and synaptic SRC phosphorylation. We propose that microtubule and actin remodeling cooperate during withdrawal to maintain the plastic structural changes initially established by cocaine self-administration. This work opens new translational avenues for further characterization of microtubule-associated regulatory molecules as putative drug targets to tackle relapse to drug taking.


Subject(s)
Cocaine/administration & dosage , Locomotion/physiology , Microtubule-Associated Proteins/metabolism , Oncogene Protein pp60(v-src)/metabolism , Substance Withdrawal Syndrome/metabolism , Synapses/metabolism , Animals , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/pathology , Female , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Microtubules/drug effects , Microtubules/metabolism , Microtubules/pathology , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Self Administration , Substance Withdrawal Syndrome/pathology , Synapses/drug effects , Synapses/pathology
2.
Neuroscience ; 353: 1-6, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28412501

ABSTRACT

Chromatin regulation, in particular ATP-dependent chromatin remodelers, have previously been shown to be important in the regulation of reward-related behaviors in animal models of mental illnesses. Here we demonstrate that BAZ1A, an accessory subunit of the ISWI family of chromatin remodeling complexes, is downregulated in the nucleus accumbens (NAc) of mice exposed repeatedly to cocaine and of cocaine-addicted humans. Viral-mediated overexpression of BAZ1A in mouse NAc reduces cocaine reward as assessed by conditioned place preference (CPP), but increases cocaine-induced locomotor activation. Furthermore, we investigate nucleosome repositioning genome-wide by conducting chromatin immunoprecipitation (ChIP)-sequencing for total H3 in NAc of control mice and after repeated cocaine administration, and find extensive nucleosome occupancy and shift changes across the genome in response to cocaine exposure. These findings implicate BAZ1A in molecular and behavioral plasticity to cocaine and offer new insight into the pathophysiology of cocaine addiction.


Subject(s)
Chromatin Assembly and Disassembly/drug effects , Chromosomal Proteins, Non-Histone/genetics , Cocaine/administration & dosage , Nucleus Accumbens/metabolism , Transcription Factors/genetics , Animals , Cocaine-Related Disorders/genetics , Conditioning, Classical/drug effects , Humans , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Nucleosomes/drug effects , Nucleosomes/metabolism , RNA, Messenger/metabolism
3.
Proc Natl Acad Sci U S A ; 113(34): 9623-8, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27506785

ABSTRACT

Repeated cocaine exposure regulates transcriptional regulation within the nucleus accumbens (NAc), and epigenetic mechanisms-such as histone acetylation and methylation on Lys residues-have been linked to these lasting actions of cocaine. In contrast to Lys methylation, the role of histone Arg (R) methylation remains underexplored in addiction models. Here we show that protein-R-methyltransferase-6 (PRMT6) and its associated histone mark, asymmetric dimethylation of R2 on histone H3 (H3R2me2a), are decreased in the NAc of mice and rats after repeated cocaine exposure, including self-administration, and in the NAc of cocaine-addicted humans. Such PRMT6 down-regulation occurs selectively in NAc medium spiny neurons (MSNs) expressing dopamine D2 receptors (D2-MSNs), with opposite regulation occurring in D1-MSNs, and serves to protect against cocaine-induced addictive-like behavioral abnormalities. Using ChIP-seq, we identified Src kinase signaling inhibitor 1 (Srcin1; also referred to as p140Cap) as a key gene target for reduced H3R2me2a binding, and found that consequent Srcin1 induction in the NAc decreases Src signaling, cocaine reward, and the motivation to self-administer cocaine. Taken together, these findings suggest that suppression of Src signaling in NAc D2-MSNs, via PRMT6 and H3R2me2a down-regulation, functions as a homeostatic brake to restrain cocaine action, and provide novel candidates for the development of treatments for cocaine addiction.


Subject(s)
Carrier Proteins/genetics , Cocaine-Related Disorders/metabolism , Cocaine/administration & dosage , Histones/metabolism , Nucleus Accumbens/metabolism , Protein Processing, Post-Translational , Acetylation , Animals , Carrier Proteins/metabolism , Cocaine-Related Disorders/genetics , Cocaine-Related Disorders/pathology , Histones/genetics , Humans , Male , Methylation , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Nucleus Accumbens/pathology , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism
4.
J Neurosci ; 36(14): 3954-61, 2016 Apr 06.
Article in English | MEDLINE | ID: mdl-27053203

ABSTRACT

ATP-dependent chromatin remodeling proteins are being implicated increasingly in the regulation of complex behaviors, including models of several psychiatric disorders. Here, we demonstrate that Baz1b, an accessory subunit of the ISWI family of chromatin remodeling complexes, is upregulated in the nucleus accumbens (NAc), a key brain reward region, in both chronic cocaine-treated mice and mice that are resilient to chronic social defeat stress. In contrast, no regulation is seen in mice that are susceptible to this chronic stress. Viral-mediated overexpression of Baz1b, along with its associated subunit Smarca5, in mouse NAc is sufficient to potentiate both rewarding responses to cocaine, including cocaine self-administration, and resilience to chronic social defeat stress. However, despite these similar, proreward behavioral effects, genome-wide mapping of BAZ1B in NAc revealed mostly distinct subsets of genes regulated by these chromatin remodeling proteins after chronic exposure to either cocaine or social stress. Together, these findings suggest important roles for BAZ1B and its associated chromatin remodeling complexes in NAc in the regulation of reward behaviors to distinct emotional stimuli and highlight the stimulus-specific nature of the actions of these regulatory proteins. SIGNIFICANCE STATEMENT: We show that BAZ1B, a component of chromatin remodeling complexes, in the nucleus accumbens regulates reward-related behaviors in response to chronic exposure to both rewarding and aversive stimuli by regulating largely distinct subsets of genes.


Subject(s)
Behavior, Animal/physiology , Emotions/physiology , Nucleus Accumbens/physiology , Reward , Transcription Factors/genetics , Transcription Factors/physiology , Adenosine Triphosphatases/metabolism , Animals , Behavior, Animal/drug effects , Chromatin/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cocaine/pharmacology , Epigenesis, Genetic/drug effects , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats , Rats, Sprague-Dawley , Self Administration , Social Environment , Stress, Psychological
5.
Nat Med ; 21(10): 1146-53, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26390241

ABSTRACT

Improved treatment for major depressive disorder (MDD) remains elusive because of the limited understanding of its underlying biological mechanisms. It is likely that stress-induced maladaptive transcriptional regulation in limbic neural circuits contributes to the development of MDD, possibly through epigenetic factors that regulate chromatin structure. We establish that persistent upregulation of the ACF (ATP-utilizing chromatin assembly and remodeling factor) ATP-dependent chromatin-remodeling complex, occurring in the nucleus accumbens of stress-susceptible mice and depressed humans, is necessary for stress-induced depressive-like behaviors. We found that altered ACF binding after chronic stress was correlated with altered nucleosome positioning, particularly around the transcription start sites of affected genes. These alterations in ACF binding and nucleosome positioning were associated with repressed expression of genes implicated in susceptibility to stress. Together, our findings identify the ACF chromatin-remodeling complex as a critical component in the development of susceptibility to depression and in regulating stress-related behaviors.


Subject(s)
Chromatin Assembly and Disassembly , Depression/metabolism , Stress, Psychological , Animals , Chromosomal Proteins, Non-Histone , Humans , Male , Mice , Mice, Inbred C57BL , Transcription Factors/genetics , Transcription Factors/physiology
6.
Nat Neurosci ; 18(7): 959-61, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26030849

ABSTRACT

Activin receptor signaling, including the transcription factor Smad3, was upregulated in the rat nucleus accumbens (NAc) shell following withdrawal from cocaine. Direct genetic and pharmacological manipulations of this pathway bidirectionally altered cocaine seeking while governing morphological plasticity in NAc neurons. Thus, Activin/Smad3 signaling is induced following withdrawal from cocaine, and such regulation may be a key molecular mechanism underlying behavioral and cellular plasticity in the brain following cocaine self-administration.


Subject(s)
Activin Receptors/metabolism , Behavior, Animal/drug effects , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Neuronal Plasticity/drug effects , Nucleus Accumbens/drug effects , Signal Transduction/drug effects , Smad3 Protein/metabolism , Animals , Cocaine/administration & dosage , Dendritic Spines/drug effects , Dopamine Uptake Inhibitors/administration & dosage , Male , Nucleus Accumbens/cytology , Rats , Rats, Sprague-Dawley , Self Administration , Signal Transduction/genetics
7.
Neurosci Lett ; 598: 23-8, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25957559

ABSTRACT

Chronic cocaine exposure increases the density of dendritic spines on medium spiny neurons (MSNs), the predominant neuronal cell type of the nucleus accumbens (NAc), a key brain reward region. We recently showed that suppression of Rac1, a small GTPase, is a critical mediator of this structural plasticity, but the upstream determinants of Rac1 activity in this context remain to be elucidated. In this study we examined whether isoforms of Dishevelled, a key hub protein of multiple branches of Wnt signaling, including Rac1, are regulated in the NAc by chronic cocaine, and whether these Dishevelled isoforms control Rac1 activity in this brain region in vivo. We found that chronic cocaine administration decreased expression of Dishevelled-2, and several other Wnt signaling components, in the NAc, and that overexpression of Dishevelled-2, but not Dishevelled-1, conversely upregulated Rac1 activity and prevented the cocaine induction of dendritic spines on NAc MSNs. We posit that the cocaine-induced downregulation of Dishevelled-2 in the NAc is an upstream regulator of Rac1 activity and plays an important role in the dynamic structural plasticity of NAc MSNs seen in response to chronic cocaine exposure.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cocaine/pharmacology , Dendritic Spines/drug effects , Nucleus Accumbens/drug effects , Phosphoproteins/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/pathology , Dendritic Spines/ultrastructure , Dishevelled Proteins , Male , Mice, Inbred C57BL , Nucleus Accumbens/metabolism , Wnt Signaling Pathway
8.
Neuron ; 86(4): 853-855, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25996128

ABSTRACT

In this issue of Neuron, innovative new modifications to opioid receptors are used to expand the tools available to modulate neuronal activity. Vardy et al. (2015) describe a new "DREADD" chemogenetic tool based on the inhibitory κ opioid receptor (KORD) that can be used in conjunction with already-available DREADDs. Siuda et al. (2015) report the development of "opto-MOR," a light-activatable µ opioid receptor (MOR) chimera that can be used to better understand the complexities of MOR signaling.


Subject(s)
Analgesics, Opioid/pharmacology , Behavior, Animal/drug effects , Diterpenes/pharmacology , GABAergic Neurons/drug effects , Neurons/drug effects , Optogenetics , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Signal Transduction/drug effects , Animals
9.
Nat Neurosci ; 18(3): 415-22, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25643298

ABSTRACT

Brain-derived neurotrophic factor (BDNF) has a crucial role in modulating neural and behavioral plasticity to drugs of abuse. We found a persistent downregulation of exon-specific Bdnf expression in the ventral tegmental area (VTA) in response to chronic opiate exposure, which was mediated by specific epigenetic modifications at the corresponding Bdnf gene promoters. Exposure to chronic morphine increased stalling of RNA polymerase II at these Bdnf promoters in VTA and altered permissive and repressive histone modifications and occupancy of their regulatory proteins at the specific promoters. Furthermore, we found that morphine suppressed binding of phospho-CREB (cAMP response element binding protein) to Bdnf promoters in VTA, which resulted from enrichment of trimethylated H3K27 at the promoters, and that decreased NURR1 (nuclear receptor related-1) expression also contributed to Bdnf repression and associated behavioral plasticity to morphine. Our findings suggest previously unknown epigenetic mechanisms of morphine-induced molecular and behavioral neuroadaptations.


Subject(s)
Analgesics, Opioid/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Epigenesis, Genetic/physiology , Ventral Tegmental Area/metabolism , Analgesics, Opioid/pharmacology , Animals , Brain-Derived Neurotrophic Factor/genetics , Cocaine/pharmacology , Conditioning, Operant/drug effects , Dopamine Uptake Inhibitors/pharmacology , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic/drug effects , Heroin Dependence/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Postmortem Changes , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Rats , Rats, Sprague-Dawley , Ventral Tegmental Area/drug effects
10.
Nature ; 516(7529): 51-5, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25383518

ABSTRACT

ß-catenin is a multi-functional protein that has an important role in the mature central nervous system; its dysfunction has been implicated in several neuropsychiatric disorders, including depression. Here we show that in mice ß-catenin mediates pro-resilient and anxiolytic effects in the nucleus accumbens, a key brain reward region, an effect mediated by D2-type medium spiny neurons. Using genome-wide ß-catenin enrichment mapping, we identify Dicer1-important in small RNA (for example, microRNA) biogenesis--as a ß-catenin target gene that mediates resilience. Small RNA profiling after excising ß-catenin from nucleus accumbens in the context of chronic stress reveals ß-catenin-dependent microRNA regulation associated with resilience. Together, these findings establish ß-catenin as a critical regulator in the development of behavioural resilience, activating a network that includes Dicer1 and downstream microRNAs. We thus present a foundation for the development of novel therapeutic targets to promote stress resilience.


Subject(s)
DEAD-box RNA Helicases/genetics , Gene Expression Regulation , MicroRNAs/genetics , Resilience, Psychological , Ribonuclease III/genetics , Stress, Physiological/genetics , beta Catenin/metabolism , Adaptation, Physiological/genetics , Animals , DEAD-box RNA Helicases/metabolism , Depression/physiopathology , Gene Expression Profiling , Genome-Wide Association Study , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Neurons/metabolism , Ribonuclease III/metabolism , Signal Transduction , beta Catenin/genetics
11.
Proc Natl Acad Sci U S A ; 111(5): 2005-10, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24449909

ABSTRACT

Many of the long-term effects of cocaine on the brain's reward circuitry have been shown to be mediated by alterations in gene expression. Several chromatin modifications, including histone acetylation and methylation, have been implicated in this regulation, but the effect of other histone modifications remains poorly understood. Poly(ADP-ribose) polymerase-1 (PARP-1), a ubiquitous and abundant nuclear protein, catalyzes the synthesis of a negatively charged polymer called poly(ADP-ribose) or PAR on histones and other substrate proteins and forms transcriptional regulatory complexes with several other chromatin proteins. Here, we identify an essential role for PARP-1 in cocaine-induced molecular, neural, and behavioral plasticity. Repeated cocaine administration, including self-administration, increased global levels of PARP-1 and its mark PAR in mouse nucleus accumbens (NAc), a key brain reward region. Using PARP-1 inhibitors and viral-mediated gene transfer, we established that PARP-1 induction in NAc mediates enhanced behavioral responses to cocaine, including increased self-administration of the drug. Using chromatin immunoprecipitation sequencing, we demonstrated a global, genome-wide enrichment of PARP-1 in NAc of cocaine-exposed mice and identified several PARP-1 target genes that could contribute to the lasting effects of cocaine. Specifically, we identified sidekick-1--important for synaptic connections during development--as a critical PARP-1 target gene involved in cocaine's behavioral effects as well as in its ability to induce dendritic spines on NAc neurons. These findings establish the involvement of PARP-1 and PARylation in the long-term actions of cocaine.


Subject(s)
Cocaine/pharmacology , Poly Adenosine Diphosphate Ribose/metabolism , Animals , Behavior, Animal/drug effects , Chromatin Immunoprecipitation , Cocaine/administration & dosage , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Genome/genetics , Immunoglobulin G/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Nucleus Accumbens/enzymology , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Substrate Specificity/drug effects , Transcription, Genetic/drug effects
12.
J Neurosci ; 33(47): 18381-95, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24259563

ABSTRACT

The transcription factor, ΔFosB, is robustly and persistently induced in striatum by several chronic stimuli, such as drugs of abuse, antipsychotic drugs, natural rewards, and stress. However, very few studies have examined the degree of ΔFosB induction in the two striatal medium spiny neuron (MSN) subtypes. We make use of fluorescent reporter BAC transgenic mice to evaluate induction of ΔFosB in dopamine receptor 1 (D1) enriched and dopamine receptor 2 (D2) enriched MSNs in ventral striatum, nucleus accumbens (NAc) shell and core, and in dorsal striatum (dStr) after chronic exposure to several drugs of abuse including cocaine, ethanol, Δ(9)-tetrahydrocannabinol, and opiates; the antipsychotic drug, haloperidol; juvenile enrichment; sucrose drinking; calorie restriction; the serotonin selective reuptake inhibitor antidepressant, fluoxetine; and social defeat stress. Our findings demonstrate that chronic exposure to many stimuli induces ΔFosB in an MSN-subtype selective pattern across all three striatal regions. To explore the circuit-mediated induction of ΔFosB in striatum, we use optogenetics to enhance activity in limbic brain regions that send synaptic inputs to NAc; these regions include the ventral tegmental area and several glutamatergic afferent regions: medial prefrontal cortex, amygdala, and ventral hippocampus. These optogenetic conditions lead to highly distinct patterns of ΔFosB induction in MSN subtypes in NAc core and shell. Together, these findings establish selective patterns of ΔFosB induction in striatal MSN subtypes in response to chronic stimuli and provide novel insight into the circuit-level mechanisms of ΔFosB induction in striatum.


Subject(s)
Corpus Striatum/cytology , Dopamine Agents/pharmacology , Emotions/drug effects , Optogenetics , Proto-Oncogene Proteins c-fos/metabolism , Animals , Antidepressive Agents/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Dronabinol/pharmacology , Environment , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/classification , Neurons/drug effects , Phosphopyruvate Hydratase/metabolism , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/genetics
13.
Nat Neurosci ; 16(4): 434-40, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23475113

ABSTRACT

Induction of histone acetylation in the nucleus accumbens (NAc), a key brain reward region, promotes cocaine-induced alterations in gene expression. Histone deacetylases (HDACs) tightly regulate the acetylation of histone tails, but little is known about the functional specificity of different HDAC isoforms in the development and maintenance of cocaine-induced plasticity, and previous studies of HDAC inhibitors report conflicting effects on cocaine-elicited behavioral adaptations. Here we demonstrate that specific and prolonged blockade of HDAC1 in NAc of mice increased global levels of histone acetylation, but also induced repressive histone methylation and antagonized cocaine-induced changes in behavior, an effect mediated in part through a chromatin-mediated suppression of GABAA receptor subunit expression and inhibitory tone on NAc neurons. Our findings suggest a new mechanism by which prolonged and selective HDAC inhibition can alter behavioral and molecular adaptations to cocaine and inform the development of therapeutics for cocaine addiction.


Subject(s)
Cocaine/pharmacology , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Animals , Benzamides/pharmacology , Cocaine/antagonists & inhibitors , Histones/antagonists & inhibitors , Histones/metabolism , Male , Methylation/drug effects , Mice , Mice, Inbred C57BL , Pyridines/pharmacology , Random Allocation
14.
J Neurosci ; 32(48): 17454-64, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23197736

ABSTRACT

Dysregulation of histone modifying enzymes has been associated with numerous psychiatric disorders. Alterations in G9a (Ehmt2), a histone methyltransferase that catalyzes the euchromatic dimethylation of histone H3 at lysine 9 (H3K9me2), has been implicated recently in mediating neural and behavioral plasticity in response to chronic cocaine administration. Here, we show that chronic morphine, like cocaine, decreases G9a expression, and global levels of H3K9me2, in mouse nucleus accumbens (NAc), a key brain reward region. In contrast, levels of other histone methyltransferases or demethylases, or of other methylated histone marks, were not affected in NAc by chronic morphine. Through viral-mediated gene transfer and conditional mutagenesis, we found that overexpression of G9a in NAc opposes morphine reward and locomotor sensitization and concomitantly promotes analgesic tolerance and naloxone-precipitated withdrawal, whereas downregulation of G9a in NAc enhances locomotor sensitization and delays the development of analgesic tolerance. We identified downstream targets of G9a by providing a comprehensive chromatin immunoprecipitation followed by massively parallel sequencing analysis of H3K9me2 distribution in NAc in the absence and presence of chronic morphine. These data provide novel insight into the epigenomic regulation of H3K9me2 by chronic morphine and suggest novel chromatin-based mechanisms through which morphine-induced addictive-like behaviors arise.


Subject(s)
Behavior, Animal/drug effects , Epigenesis, Genetic/drug effects , Histones/genetics , Morphine/pharmacology , Narcotics/pharmacology , Nucleus Accumbens/drug effects , Animals , DNA Methylation/drug effects , Gene Transfer Techniques , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Morphine/adverse effects , Motor Activity/drug effects , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Narcotics/adverse effects , Nucleus Accumbens/metabolism , Substance Withdrawal Syndrome/metabolism
15.
Science ; 338(6103): 124-8, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-23042896

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a key positive regulator of neural plasticity, promoting, for example, the actions of stimulant drugs of abuse such as cocaine. We discovered a surprising opposite role for BDNF in countering responses to chronic morphine exposure. The suppression of BDNF in the ventral tegmental area (VTA) enhanced the ability of morphine to increase dopamine (DA) neuron excitability and promote reward. In contrast, optical stimulation of VTA DA terminals in nucleus accumbens (NAc) completely reversed the suppressive effect of BDNF on morphine reward. Furthermore, we identified numerous genes in the NAc, a major target region of VTA DA neurons, whose regulation by BDNF in the context of chronic morphine exposure mediated this counteractive function. These findings provide insight into the molecular basis of morphine-induced neuroadaptations in the brain's reward circuitry.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Dopaminergic Neurons/drug effects , Morphine Dependence/physiopathology , Morphine/pharmacology , Ventral Tegmental Area/drug effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Dopamine/metabolism , Dopaminergic Neurons/physiology , Gene Expression Regulation , Gene Knockdown Techniques , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL , Morphine Dependence/genetics , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiopathology , Photic Stimulation , Receptor, trkB/genetics , Receptor, trkB/physiology , Ventral Tegmental Area/physiology
16.
J Neurosci ; 32(30): 10267-72, 2012 Jul 25.
Article in English | MEDLINE | ID: mdl-22836260

ABSTRACT

ΔFosB, a Fosb gene product, is induced in nucleus accumbens (NAc) and caudate-putamen (CPu) by repeated exposure to drugs of abuse such as cocaine. This induction contributes to aberrant patterns of gene expression and behavioral abnormalities seen with repeated drug exposure. Here, we assessed whether a remote history of cocaine exposure in rats might alter inducibility of the Fosb gene elicited by subsequent drug exposure. We show that prior chronic cocaine administration, followed by extended withdrawal, increases inducibility of Fosb in NAc, as evidenced by greater acute induction of ΔFosB mRNA and faster accumulation of ΔFosB protein after repeated cocaine reexposure. No such primed Fosb induction was observed in CPu; in fact, subsequent acute induction of ΔFosB mRNA was suppressed in CPu. These abnormal patterns of Fosb expression are associated with chromatin modifications at the Fosb gene promoter. Prior chronic cocaine administration induces a long-lasting increase in RNA polymerase II (Pol II) binding at the Fosb promoter in NAc only, suggesting that Pol II "stalling" primes Fosb for induction in this region upon reexposure to cocaine. A cocaine challenge then triggers the release of Pol II from the gene promoter, allowing for more rapid Fosb transcription. A cocaine challenge also decreases repressive histone modifications at the Fosb promoter in NAc, but increases such repressive marks and decreases activating marks in CPu. These results provide new insight into the chromatin dynamics at the Fosb promoter and reveal a novel mechanism for primed Fosb induction in NAc upon reexposure to cocaine.


Subject(s)
Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Nucleus Accumbens/drug effects , Proto-Oncogene Proteins c-fos/genetics , Animals , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Gene Expression/drug effects , Male , Motor Activity/drug effects , Nucleus Accumbens/metabolism , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley
17.
Nat Neurosci ; 15(6): 891-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22522400

ABSTRACT

Repeated cocaine administration increases the dendritic arborization of nucleus accumbens neurons, but the underlying signaling events remain unknown. Here we show that repeated exposure to cocaine negatively regulates the active form of Rac1, a small GTPase that controls actin remodeling in other systems. Further, we show, using viral-mediated gene transfer, that overexpression of a dominant negative mutant of Rac1 or local knockout of Rac1 is sufficient to increase the density of immature dendritic spines on nucleus accumbens neurons, whereas overexpression of a constitutively active Rac1 or light activation of a photoactivatable form of Rac1 blocks the ability of repeated cocaine exposure to produce this effect. Downregulation of Rac1 activity likewise promotes behavioral responses to cocaine exposure, with activation of Rac1 producing the opposite effect. These findings establish that Rac1 signaling mediates structural and behavioral plasticity in response to cocaine exposure.


Subject(s)
Cocaine/pharmacology , Dendritic Spines/drug effects , Dopamine Uptake Inhibitors/pharmacology , Neuronal Plasticity/drug effects , Neuropeptides/metabolism , Signal Transduction/drug effects , rac GTP-Binding Proteins/metabolism , Animals , Blotting, Western , Cocaine-Related Disorders , Dendritic Spines/metabolism , Gene Knockdown Techniques , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Neurons/drug effects , Neurons/metabolism , Neuropeptides/genetics , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein
18.
Stem Cells Int ; 2011: 586586, 2011.
Article in English | MEDLINE | ID: mdl-22190964

ABSTRACT

Bone marrow-derived mesenchymal stromal cells (MSCs) hold promise for autologous treatment of neuropathologies. Intranasal delivery is relatively noninvasive and has recently been reported to result in transport of MSCs to the brain. However, the ability of MSCs to migrate from nasal passages to sites of neuropathology and ultimately survive has not been fully examined. In this paper, we harvested MSCs from transgenic mice expressing enhanced green fluorescent protein (cells hereafter referred to as MSC-EGFP) and delivered them intranasally to wild-type mice sustaining mechanical lesions in the striatum. Using fluorescent, colorimetric, and ultrastructural detection methods, GFP-expressing cells were undetectable in the brain from 3 hours to 2 months after MSC delivery. However, bright autofluorescence that strongly resembled emission from GFP was observed in the olfactory bulb and striatum of lesioned control and MSC-EGFP-treated mice. In a control experiment, we directly implanted MSC-EGFPs into the mouse striatum and detected robust GFP expression 1 and 7 days after implantation. These findings suggest that-under our conditions-intranasally delivered MSC-EGFPs do not survive or migrate in the brain. Furthermore, our observations highlight the necessity of including appropriate controls when working with GFP as a cellular marker.

19.
Biol Psychiatry ; 70(8): 744-753, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21757186

ABSTRACT

BACKGROUND: Repeated exposure to drugs of abuse and stress increase dynorphin, a κ opioid receptor (KOR) ligand, in the nucleus accumbens (NAc). Acute KOR activation produces dysphoria that might contribute to addictive behavior. How repeated KOR activation modulates reward circuitry is not understood. METHODS: We used intracranial self-stimulation (ICSS), a method that provides a behavioral index of reward sensitivity, to measure the effects of repeated administration of the KOR agonist salvinorin A (salvA) (2 mg/kg) on the reward-potentiating effects of cocaine (5.0 mg/kg). In separate rats, we measured the effects of salvA on activation of extracellular signal regulated kinase (ERK), cyclic adenosine monophosphate (cAMP) response element binding protein, and c-Fos within the NAc. RESULTS: SalvA had biphasic effects on reward: an immediate effect was to decrease the rewarding impact of ICSS, whereas a delayed effect was to increase the rewarding impact of ICSS. Repeated salvA produced a net decrease in the reward-potentiating effects of cocaine. In the NAc, both acute and repeated salvA administration increased phosphorylated ERK, whereas only acute salvA increased c-Fos and repeated salvA increased phosphorylated cAMP response element binding protein. The KOR antagonist nor-binaltorphimine (20 mg/kg) blocked the immediate and delayed effects of salvA and prolonged the duration of cocaine effects in ICSS. CONCLUSIONS: Repeated salvA might trigger opponent processes such that "withdrawal" from the dysphoric effects of KOR activation is rewarding and decreases the net rewarding valence of cocaine. The temporal effects of salvA on ERK signaling suggest KOR-mediated engagement of distinct signaling pathways within the NAc that might contribute to biphasic effects on reward sensitivity.


Subject(s)
Diterpenes, Clerodane/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, Opioid, kappa/agonists , Reward , Self Stimulation/drug effects , Animals , Cocaine/antagonists & inhibitors , Cocaine/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Diterpenes, Clerodane/antagonists & inhibitors , Drug Interactions/physiology , Male , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Nucleus Accumbens/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/antagonists & inhibitors , Self Stimulation/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Time Factors
20.
Science ; 330(6002): 385-90, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20947769

ABSTRACT

The nucleus accumbens is a key mediator of cocaine reward, but the distinct roles of the two subpopulations of nucleus accumbens projection neurons, those expressing dopamine D1 versus D2 receptors, are poorly understood. We show that deletion of TrkB, the brain-derived neurotrophic factor (BDNF) receptor, selectively from D1+ or D2+ neurons oppositely affects cocaine reward. Because loss of TrkB in D2+ neurons increases their neuronal excitability, we next used optogenetic tools to control selectively the firing rate of D1+ and D2+ nucleus accumbens neurons and studied consequent effects on cocaine reward. Activation of D2+ neurons, mimicking the loss of TrkB, suppresses cocaine reward, with opposite effects induced by activation of D1+ neurons. These results provide insight into the molecular control of D1+ and D2+ neuronal activity as well as the circuit-level contribution of these cell types to cocaine reward.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cocaine-Related Disorders/metabolism , Cocaine/pharmacology , Neurons/metabolism , Nucleus Accumbens/metabolism , Receptor, trkB/metabolism , Reward , Signal Transduction , Animals , Behavior, Animal/drug effects , Channelrhodopsins , Conditioning, Psychological , Light , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , Motor Activity/drug effects , Nucleus Accumbens/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, trkB/genetics , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...