Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Addict Biol ; 26(1): e12816, 2021 01.
Article in English | MEDLINE | ID: mdl-31373129

ABSTRACT

Epigenetic enzymes oversee long-term changes in gene expression by integrating genetic and environmental cues. While there are hundreds of enzymes that control histone and DNA modifications, their potential roles in substance abuse and alcohol dependence remain underexplored. A few recent studies have suggested that epigenetic processes could underlie transcriptomic and behavioral hallmarks of alcohol addiction. In the present study, we sought to identify epigenetic enzymes in the brain that are dysregulated during protracted abstinence as a consequence of chronic and intermittent alcohol exposure. Through quantitative mRNA expression analysis of over 100 epigenetic enzymes, we identified 11 that are significantly altered in alcohol-dependent rats compared with controls. Follow-up studies of one of these enzymes, the histone demethylase KDM6B, showed that this enzyme exhibits region-specific dysregulation in the prefrontal cortex and nucleus accumbens of alcohol-dependent rats. KDM6B was also upregulated in the human alcoholic brain. Upregulation of KDM6B protein in alcohol-dependent rats was accompanied by a decrease of trimethylation levels at histone H3, lysine 27 (H3K27me3), consistent with the known demethylase specificity of KDM6B. Subsequent epigenetic (chromatin immunoprecipitation [ChIP]-sequencing) analysis showed that alcohol-induced changes in H3K27me3 were significantly enriched at genes in the IL-6 signaling pathway, consistent with the well-characterized role of KDM6B in modulation of inflammatory responses. Knockdown of KDM6B in cultured microglial cells diminished IL-6 induction in response to an inflammatory stimulus. Our findings implicate a novel KDM6B-mediated epigenetic signaling pathway integrated with inflammatory signaling pathways that are known to underlie the development of alcohol addiction.


Subject(s)
Alcoholism/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Animals , Cells, Cultured , Epigenesis, Genetic , Ethanol/metabolism , Histone Demethylases/genetics , Histones/metabolism , Humans , Prefrontal Cortex/metabolism , Rats , Signal Transduction , Up-Regulation
2.
Pharmacol Biochem Behav ; 196: 172973, 2020 09.
Article in English | MEDLINE | ID: mdl-32569786

ABSTRACT

Early life trauma dramatically increases the risk of developing major depressive disorder (MDD), and is associated with a markedly decreased adult treatment response to antidepressants. Novel treatment approaches are required to treat childhood trauma-associated MDD. Recent studies suggest that the (R,S)-ketamine (ketamine) metabolite, (2R,6R)-hydroxynorketamine (HNK), exerts fast- and long-lasting antidepressant-like effects without ketamine's NMDAR-inhibition-associated adverse side-effect profile. We investigated the therapeutic potential of (2R,6R)-HNK against behavioral despair produced by a novel live-predator stress exposure during adolescence. Male and female C57BL/6J mice were exposed to a live snake or control conditions at post-natal (PND) days 31, 45 and 61. In order to assess the enduring consequences of trauma-exposure, at a minimum of 14 days following the last exposure, mice received inescapable shocks followed by a session with available escape options twenty-four hours later. Mice that manifested enduring escape deficits (helplessness) were treated with vehicle or (2R,6R)-HNK (20 mg/kg, i.p.), 24 h prior to retesting for reversal of escape deficits. We found that a significantly greater number of mice developed the helpless phenotype when they were exposed to the live predator and that the helpless phenotype was reversed in mice treated with (2R,6R)-HNK. There were no sex differences in the response to predator-stress exposure or (2R,6R)-HNK treatment. The live-predator model developed in this study provides an opportunity to further refine our understanding of the neurobiological substrates impacted by adolescent trauma and improve treatment strategies. The demonstrated efficacy of (2R,6R)-HNK in this model suggests a novel therapeutic intervention for a treatment-resistant population.


Subject(s)
Behavior, Animal/drug effects , Depressive Disorder, Major/drug therapy , Ketamine/analogs & derivatives , Animals , Female , Ketamine/therapeutic use , Male , Mice , Mice, Inbred C57BL , Predatory Behavior , Stress, Psychological
3.
Mol Psychiatry ; 25(2): 461-475, 2020 02.
Article in English | MEDLINE | ID: mdl-29728704

ABSTRACT

Rodent studies indicate that ghrelin receptor blockade reduces alcohol consumption. However, no ghrelin receptor blockers have been administered to heavy alcohol drinking individuals. Therefore, we evaluated the safety, tolerability, pharmacokinetic (PK), pharmacodynamic (PD) and behavioral effects of a novel ghrelin receptor inverse agonist, PF-5190457, when co-administered with alcohol. We tested the effects of PF-5190457 combined with alcohol on locomotor activity, loss-of-righting reflex (a measure of alcohol sedative actions), and on blood PF-5190457 concentrations in rats. Then, we performed a single-blind, placebo-controlled, within-subject human study with PF-5190457 (placebo/0 mg b.i.d., 50 mg b.i.d., 100 mg b.i.d.). Twelve heavy drinkers during three identical visits completed an alcohol administration session, subjective assessments, and an alcohol cue-reactivity procedure, and gave blood samples for PK/PD testing. In rats, PF-5190457 did not interact with the effects of alcohol on locomotor activity or loss-of-righting reflex. Alcohol did not affect blood PF-5190457 concentrations. In humans, all adverse events were mild or moderate and did not require discontinuation or dose reductions. Drug dose did not alter alcohol concentration or elimination, alcohol-induced stimulation or sedation, or mood during alcohol administration. Potential PD markers of PF-5190457 were acyl-to-total ghrelin ratio and insulin-like growth factor-1. PF-5190457 (100 mg b.i.d.) reduced alcohol craving during the cue-reactivity procedure. This study provides the first translational evidence of safety and tolerability of the ghrelin receptor inverse agonist PF-5190457 when co-administered with alcohol. PK/PD/behavioral findings support continued research of PF-5190457 as a potential pharmacological agent to treat alcohol use disorder.


Subject(s)
Alcoholic Intoxication/drug therapy , Azetidines/pharmacology , Receptors, Ghrelin/agonists , Spiro Compounds/pharmacology , Adult , Alcohol Drinking/drug therapy , Alcohol Drinking/metabolism , Alcoholism/drug therapy , Alcoholism/metabolism , Animals , Azetidines/metabolism , Azetidines/pharmacokinetics , Ethanol/chemistry , Female , Ghrelin/metabolism , Humans , Male , Middle Aged , Rats , Rats, Wistar , Receptors, Ghrelin/metabolism , Research Design , Single-Blind Method , Spiro Compounds/metabolism , Spiro Compounds/pharmacokinetics
4.
Addict Biol ; 22(5): 1279-1288, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27273552

ABSTRACT

Proinflammatory activity has been postulated to play a role in addictive processes and stress responses, but the underlying mechanisms remain largely unknown. Here, we examined the role of interleukin 1 (IL-1) and tumor necrosis factor-α (TNF-α) in regulation of voluntary alcohol consumption, alcohol reward and stress-induced drinking. Mice with a deletion of the IL-1 receptor I gene (IL-1RI KO) exhibited modestly decreased alcohol consumption. However, IL-1RI deletion affected neither the rewarding properties of alcohol, measured by conditioned place preference (CPP), nor stress-induced drinking induced by social defeat stress. TNF-α signaling can compensate for phenotypic consequences of IL1-RI deletion. We therefore hypothesized that double deletion of both IL-1RI and TNF-1 receptors (TNF-1R) may reveal the role of these pathways in regulation of alcohol intake. Double KOs consumed significantly less alcohol than control mice over a range of alcohol concentrations. The combined deletion of TNF-1R and IL-1RI did not influence alcohol reward, but did prevent increased alcohol consumption resulting from exposure to repeated bouts of social defeat stress. Taken together, these data indicate that IL-1RI and TNF-1R contribute to regulation of stress-induced, negatively reinforced drinking perhaps through overlapping signaling events downstream of these receptors, while leaving rewarding properties of alcohol largely unaffected.


Subject(s)
Alcohol Drinking/immunology , Behavior, Animal , Interleukin-1/immunology , Receptors, Interleukin-1 Type I/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Stress, Psychological/immunology , Tumor Necrosis Factor-alpha/immunology , Alcohol Drinking/genetics , Animals , Central Nervous System Depressants/administration & dosage , Conditioning, Classical , Ethanol/administration & dosage , Inflammation , Male , Mice , Mice, Knockout , Psychological Distance , Receptors, Interleukin-1 Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Signal Transduction , Stress, Psychological/genetics
5.
Front Mol Neurosci ; 9: 20, 2016.
Article in English | MEDLINE | ID: mdl-27047334

ABSTRACT

Long-term opioid treatment results in reduced therapeutic efficacy and in turn leads to an increase in the dose required to produce equivalent pain relief and alleviate break-through or insurmountable pain. Altered gene expression is a likely means for inducing long-term neuroadaptations responsible for tolerance. Studies conducted by our laboratory (Tapocik et al., 2009) revealed a network of gene expression changes occurring in canonical pathways involved in neuroplasticity, and uncovered miRNA processing as a potential mechanism. In particular, the mRNA coding the protein responsible for processing miRNAs, Dicer1, was positively correlated with the development of analgesic tolerance. The purpose of the present study was to test the hypothesis that miRNAs play a significant role in the development of analgesic tolerance as measured by thermal nociception. Dicer1 knockdown, miRNA profiling, bioinformatics, and confirmation of high value targets were used to test the proposition. Regionally targeted Dicer1 knockdown (via shRNA) had the anticipated consequence of eliminating the development of tolerance in C57BL/6J (B6) mice, thus supporting the involvement of miRNAs in the development of tolerance. MiRNA expression profiling identified a core set of chronic morphine-regulated miRNAs (miR's 27a, 9, 483, 505, 146b, 202). Bioinformatics approaches were implemented to identify and prioritize their predicted target mRNAs. We focused our attention on miR27a and its predicted target serpin peptidase inhibitor clade I (Serpini1) mRNA, a transcript known to be intricately involved in dendritic spine density regulation in a manner consistent with chronic morphine's consequences and previously found to be correlated with the development of analgesic tolerance. In vitro reporter assay confirmed the targeting of the Serpini1 3'-untranslated region by miR27a. Interestingly miR27a was found to positively regulate Serpini1 mRNA and protein levels in multiple neuronal cell lines. Lastly, Serpini1 knockout mice developed analgesic tolerance at a slower rate than wild-type mice thus confirming a role for the protein in analgesic tolerance. Overall, these results provide evidence to support a specific role for miR27a and Serpini1 in the behavioral response to chronic opioid administration (COA) and suggest that miRNA expression and mRNA targeting may underlie the neuroadaptations that mediate tolerance to the analgesic effects of morphine.

6.
J Neurosci ; 35(15): 6153-64, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25878287

ABSTRACT

Recent studies have suggested an association between alcoholism and DNA methylation, a mechanism that can mediate long-lasting changes in gene transcription. Here, we examined the contribution of DNA methylation to the long-term behavioral and molecular changes induced by a history of alcohol dependence. In search of mechanisms underlying persistent rather than acute dependence-induced neuroadaptations, we studied the role of DNA methylation regulating medial prefrontal cortex (mPFC) gene expression and alcohol-related behaviors in rats 3 weeks into abstinence following alcohol dependence. Postdependent rats showed escalated alcohol intake, which was associated with increased DNA methylation as well as decreased expression of genes encoding synaptic proteins involved in neurotransmitter release in the mPFC. Infusion of the DNA methyltransferase inhibitor RG108 prevented both escalation of alcohol consumption and dependence-induced downregulation of 4 of the 7 transcripts modified in postdependent rats. Specifically, RG108 treatment directly reversed both downregulation of synaptotagmin 2 (Syt2) gene expression and hypermethylation on CpG#5 of its first exon. Lentiviral inhibition of Syt2 expression in the mPFC increased aversion-resistant alcohol drinking, supporting a mechanistic role of Syt2 in compulsive-like behavior. Our findings identified a functional role of DNA methylation in alcohol dependence-like behavioral phenotypes and a candidate gene network that may mediate its effects. Together, these data provide novel evidence for DNA methyltransferases as potential therapeutic targets in alcoholism.


Subject(s)
Alcoholism/pathology , Alcoholism/physiopathology , DNA Methylation/physiology , Neuronal Plasticity/physiology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Animals , Choice Behavior , Conditioning, Operant , DNA Methylation/drug effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Ethanol/administration & dosage , Gene Expression Profiling , Male , Methyltransferases/metabolism , Nerve Tissue Proteins/metabolism , Phthalimides/pharmacology , Rats , Rats, Wistar , Self Administration , Statistics, Nonparametric , Synaptotagmin II/genetics , Synaptotagmin II/metabolism , Transduction, Genetic , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
8.
Neuropsychopharmacology ; 40(4): 927-37, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25311134

ABSTRACT

PPARγ is one of the three isoforms identified for the peroxisome proliferator-activated receptors (PPARs) and is the receptor for the thiazolidinedione class of anti-diabetic medications including pioglitazone. PPARγ has been long studied for its role in adipogenesis and glucose metabolism, but the discovery of the localization in ventral tegmental area (VTA) neurons opens new vistas for a potential role in the regulation of reward processing and motivated behavior in drug addiction. Here, we demonstrate that activation of PPARγ by pioglitazone reduces the motivation for heroin and attenuates its rewarding properties. These effects are associated with a marked reduction of heroin-induced increase in phosphorylation of DARPP-32 protein in the nucleus accumbens (NAc) and with a marked and selective reduction of acute heroin-induced elevation of extracellular dopamine (DA) levels in the NAc shell, as measured by in vivo microdialysis. Through ex vivo electrophysiology in acute midbrain slices, we also show that stimulation of PPARγ attenuates opioid-induced excitation of VTA DA neurons via reduction of presynaptic GABA release from the rostromedial tegmental nucleus (RMTg). Consistent with this finding, site-specific microinjection of pioglitazone into the RMTg but not into the VTA reduced heroin taking. Our data illustrate that activation of PPARγ may represent a new pharmacotherapeutic option for the treatment of opioid addiction.


Subject(s)
Dopamine/metabolism , Heroin/administration & dosage , Narcotics/administration & dosage , Nucleus Accumbens/drug effects , PPAR gamma/metabolism , Synaptic Transmission/physiology , Anilides/pharmacology , Animals , Conditioning, Operant/drug effects , Dopaminergic Neurons/drug effects , Hypoglycemic Agents/pharmacology , Male , Mice, Transgenic , Morphine/pharmacology , PPAR gamma/genetics , Pioglitazone , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Rats , Rats, Wistar , Self Administration , Synaptic Transmission/drug effects , Thiazolidinediones/pharmacology , Time Factors , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , gamma-Aminobutyric Acid/pharmacology
9.
Neuropsychopharmacology ; 39(8): 2029-38, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24603855

ABSTRACT

Alcohol withdrawal is associated with hypothalamic-pituitary-adrenal (HPA) axis dysfunction. The FKBP5 gene codes for a co-chaperone, FK506-binding protein 5, that exerts negative feedback on HPA axis function. This study aimed to examine the effects of single-nucleotide polymorphisms (SNPs) of the FKBP5 gene in humans and the effect of Fkbp5 gene deletion in mice on alcohol withdrawal severity. We genotyped six FKBP5 SNPs (rs3800373, rs9296158, rs3777747, rs9380524, rs1360780, and rs9470080) in 399 alcohol-dependent inpatients with alcohol consumption 48 h before admission and recorded scores from the Clinical Institute Withdrawal Assessment-Alcohol revised (CIWA-Ar). Fkbp5 gene knockout (KO) and wild-type (WT) mice were assessed for alcohol withdrawal using handling-induced convulsions (HICs) following both acute and chronic alcohol exposure. We found the minor alleles of rs3800373 (G), rs9296158 (A), rs1360780 (T), and rs9470080 (T) were significantly associated with lower CIWA-Ar scores whereas the minor alleles of rs3777747 (G) and rs9380524 (A) were associated with higher scores. The haplotype-based analyses also showed an association with alcohol withdrawal severity. Fkbp5 KO mice showed significantly greater HICs during withdrawal from chronic alcohol exposure compared with WT controls. This study is the first to show a genetic effect of FKBP5 on the severity of alcohol withdrawal syndrome. In mice, the absence of the Fkbp5 gene enhances sensitivity to alcohol withdrawal. We suggest that FKBP5 variants may trigger different adaptive changes in HPA axis regulation during alcohol withdrawal with concomitant effects on withdrawal severity.


Subject(s)
Ethanol/adverse effects , Substance Withdrawal Syndrome/genetics , Tacrolimus Binding Proteins/genetics , Adult , Animals , Female , Genetic Association Studies , Haplotypes , Humans , Male , Mice , Mice, Knockout , Polymorphism, Single Nucleotide , Severity of Illness Index
10.
J Neurosci ; 34(13): 4581-8, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24672003

ABSTRACT

Escalation of voluntary alcohol consumption is a hallmark of alcoholism, but its neural substrates remain unknown. In rats, escalation occurs following prolonged exposure to cycles of alcohol intoxication, and is associated with persistent, wide-ranging changes in gene expression within the medial prefrontal cortex (mPFC). Here, we examined whether induction of microRNA (miR) 206 in mPFC contributes to escalated alcohol consumption. Following up on a microarray screen, quantitative real-time reverse transcription PCR (qPCR) confirmed that a history of dependence results in persistent (>3weeks) up-regulation of miR-206 expression in the mPFC, but not in the ventral tegmental area, amygdala, or nucleus accumbens. Viral-mediated overexpression of miR-206 in the mPFC of nondependent rats reproduced the escalation of alcohol self-administration seen following a history of dependence and significantly inhibited BDNF expression. Bioinformatic analysis identified three conserved target sites for miR-206 in the 3'-UTR of the rat BDNF transcript. Accordingly, BDNF was downregulated in post-dependent rats on microarray analysis, and this was confirmed by qPCR. In vitro, BDNF expression was repressed by miR-206 but not miR-9 in a 3'-UTR reporter assay, confirming BDNF as a functional target of miR-206. Mutation analysis showed that repression was dependent on the presence of all three miR-206 target sites in the BDNF 3'-UTR. Inhibition of miR-206 expression in differentiated rat cortical primary neurons significantly increased secreted levels of BDNF. In conclusion, recruitment of miR-206 in the mPFC contributes to escalated alcohol consumption following a history of dependence, with BDNF as a possible mediator of its action.


Subject(s)
Alcohol Drinking/pathology , Brain-Derived Neurotrophic Factor/metabolism , Gene Expression Regulation/physiology , MicroRNAs/metabolism , Prefrontal Cortex/metabolism , Alcohol Drinking/blood , Alcohols/administration & dosage , Alcohols/blood , Analysis of Variance , Animals , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , MicroRNAs/genetics , Mutation/genetics , Neurons/metabolism , Prefrontal Cortex/cytology , Rats , Rats, Wistar , Self Administration , Time Factors , Transduction, Genetic
11.
Proc Natl Acad Sci U S A ; 110(42): 16963-8, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24082084

ABSTRACT

Identification of genes influencing complex traits is hampered by genetic heterogeneity, the modest effect size of many alleles, and the likely involvement of rare and uncommon alleles. Etiologic complexity can be simplified in model organisms. By genomic sequencing, linkage analysis, and functional validation, we identified that genetic variation of Grm2, which encodes metabotropic glutamate receptor 2 (mGluR2), alters alcohol preference in animal models. Selectively bred alcohol-preferring (P) rats are homozygous for a Grm2 stop codon (Grm2 *407) that leads to largely uncompensated loss of mGluR2. mGluR2 receptor expression was absent, synaptic glutamate transmission was impaired, and expression of genes involved in synaptic function was altered. Grm2 *407 was linked to increased alcohol consumption and preference in F2 rats generated by intercrossing inbred P and nonpreferring rats. Pharmacologic blockade of mGluR2 escalated alcohol self-administration in Wistar rats, the parental strain of P and nonpreferring rats. The causal role of mGluR2 in altered alcohol preference was further supported by elevated alcohol consumption in Grm2 (-/-) mice. Together, these data point to mGluR2 as an origin of alcohol preference and a potential therapeutic target.


Subject(s)
Alcohol Drinking/genetics , Codon, Terminator , Receptors, Metabotropic Glutamate , Synaptic Transmission/genetics , Alcohol Drinking/drug therapy , Alcohol Drinking/metabolism , Alcohol Drinking/pathology , Animals , Crosses, Genetic , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glutamic Acid/metabolism , Mice , Mice, Knockout , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/biosynthesis , Receptors, Metabotropic Glutamate/genetics , Synapses/genetics , Synapses/metabolism , Synaptic Transmission/drug effects
12.
J Neurosci ; 33(24): 10132-42, 2013 Jun 12.
Article in English | MEDLINE | ID: mdl-23761908

ABSTRACT

The Neuropeptide S receptor, a Gs/Gq-coupled GPCR expressed in brain regions involved in mediating drug reward, has recently emerged as a candidate therapeutic target in addictive disorders. Here, we describe the in vitro and in vivo pharmacology of a novel, selective and brain penetrant NPSR antagonist with nanomolar affinity for the NPSR, NCGC00185684. In vitro, NCGC00185684 shows biased antagonist properties, and preferentially blocks ERK-phosphorylation over intracellular cAMP or calcium responses to NPS. In vivo, systemic NCGC00185684 blocks alcohol-induced ERK-phosphorylation in the rat central amygdala, a region involved in regulation of alcohol intake. NCGC00185684 also decreases operant alcohol self-administration, and lowers motivation for alcohol reward as measured using progressive ratio responding. These effects are behaviorally specific, in that they are observed at doses that do not influence locomotor activity or reinstatement responding following extinction. Together, these data provide an initial validation of the NPSR as a therapeutic target in alcoholism.


Subject(s)
Amygdala/drug effects , Central Nervous System Depressants/administration & dosage , Conditioning, Operant/drug effects , Ethanol/administration & dosage , Mitogen-Activated Protein Kinases/metabolism , Receptors, Neuropeptide/antagonists & inhibitors , Analysis of Variance , Animals , Cricetinae , Cricetulus , Cues , Drug Interactions , Fluorescence Resonance Energy Transfer , Humans , Imidazoles/pharmacology , In Vitro Techniques , Locomotion/drug effects , Male , Organothiophosphorus Compounds/pharmacology , Phosphorylation/drug effects , Protein Binding/drug effects , Radioligand Assay , Rats , Rats, Wistar , Receptors, Neuropeptide/metabolism , Reflex/drug effects , Reinforcement Schedule , Reinforcement, Psychology , Saccharin/administration & dosage , Self Administration , Sweetening Agents/administration & dosage , Transfection
13.
Biol Psychiatry ; 73(8): 774-81, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23419547

ABSTRACT

BACKGROUND: Genetic deletion or antagonism of the neurokinin 1 receptor (NK1R) decreases alcohol intake, alcohol reward, and stress-induced alcohol relapse in rodents, while TACR1 variation is associated with alcoholism in humans. METHODS: We used L822429, a specific antagonist with high affinity for the rat NK1R, and examined whether sensitivity to NK1R blockade is altered in alcohol-preferring (P) rats. Operant alcohol self-administration and progressive ratio responding were analyzed in P-rats and their founder Wistar line. We also analyzed Tacr1 expression and binding and sequenced the Tacr1 promoter from both lines. RESULTS: Systemic L822429 decreased alcohol self-administration in P-rats but did not affect the lower rates of alcohol self-administration in Wistar rats. Tacr1 expression was elevated in the prefrontal cortex and the amygdala of P-rats. In central amygdala, elevated Tacr1 expression was accompanied by elevated NK1R binding. Central amygdala (but not prefrontal cortex) infusion of L822429 replicated the systemic antagonist effects on alcohol self-administration in P-rats. All P-rats, but only 18% of their founder Wistar population, were CC homozygous for a-1372G/C single nucleotide polymorphism. In silico analysis indicated that the Tacr1-1372 genotype could modulate binding of the transcription factors GATA-2 and E2F-1. Electromobility shift and luciferase reporter assays suggested that the-1372C allele confers increased transcription factor binding and transcription. CONCLUSIONS: Genetic variation at the Tacr1 locus may contribute to elevated rates of alcohol self-administration, while at the same time increasing sensitivity to NK1R antagonist treatment.


Subject(s)
Alcohol Drinking/genetics , Ethanol/pharmacology , Neurokinin-1 Receptor Antagonists/pharmacology , Polymorphism, Single Nucleotide/genetics , Receptors, Neurokinin-1/metabolism , Amygdala/drug effects , Amygdala/metabolism , Animals , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , E2F1 Transcription Factor/metabolism , Ethanol/administration & dosage , GATA Transcription Factors/metabolism , Male , Microinjections , Neurokinin-1 Receptor Antagonists/administration & dosage , Piperidines/administration & dosage , Piperidines/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Receptors, Neurokinin-1/genetics , Self Administration
14.
Addict Biol ; 18(3): 480-95, 2013 May.
Article in English | MEDLINE | ID: mdl-22804800

ABSTRACT

Neuroadaptations in the ventral striatum (VS) and ventral midbrain (VMB) following chronic opioid administration are thought to contribute to the pathogenesis and persistence of opiate addiction. In order to identify candidate genes involved in these neuroadaptations, we utilized a behavior-genetics strategy designed to associate contingent intravenous drug self-administration with specific patterns of gene expression in inbred mice differentially predisposed to the rewarding effects of morphine. In a Yoked-control paradigm, C57BL/6J mice showed clear morphine-reinforced behavior, whereas DBA/2J mice did not. Moreover, the Yoked-control paradigm revealed the powerful consequences of self-administration versus passive administration at the level of gene expression. Morphine self-administration in the C57BL/6J mice uniquely up- or down-regulated 237 genes in the VS and 131 genes in the VMB. Interestingly, only a handful of the C57BL/6J self-administration genes (<3%) exhibited a similar expression pattern in the DBA/2J mice. Hence, specific sets of genes could be confidently assigned to regional effects of morphine in a contingent- and genotype-dependent manner. Bioinformatics analysis revealed that neuroplasticity, axonal guidance and micro-RNAs (miRNAs) were among the key themes associated with drug self-administration. Noteworthy were the primary miRNA genes H19 and micro-RNA containing gene (Mirg), processed, respectively, to mature miRNAs miR-675 and miR-154, because they are prime candidates to mediate network-like changes in responses to chronic drug administration. These miRNAs have postulated roles in dopaminergic neuron differentiation and mu-opioid receptor regulation. The strategic approach designed to focus on reinforcement-associated genes provides new insight into the role of neuroplasticity pathways and miRNAs in drug addiction.


Subject(s)
MicroRNAs/genetics , Morphine Dependence/genetics , Morphine/pharmacology , Narcotics/pharmacology , Neuronal Plasticity/drug effects , Adaptation, Physiological , Analysis of Variance , Animals , Axons/drug effects , Infusions, Intravenous , Mice , Mice, Inbred C57BL , Microarray Analysis , Reinforcement, Psychology , Reward , Self Administration
15.
J Neurosci ; 29(16): 5295-307, 2009 Apr 22.
Article in English | MEDLINE | ID: mdl-19386926

ABSTRACT

Chronic morphine administration may alter the expression of hundreds to thousands of genes. However, only a subset of these genes is likely involved in analgesic tolerance. In this report, we used a behavior genetics strategy to identify candidate genes specifically linked to the development of morphine tolerance. Two inbred genotypes [C57BL/6J (B6), DBA2/J (D2)] and two reciprocal congenic genotypes (B6D2, D2B6) with the proximal region of chromosome 10 (Chr10) introgressed into opposing backgrounds served as the behavior genetic filter. Tolerance after therapeutically relevant doses of morphine developed most rapidly in the B6 followed by the B6D2 genotype and did not develop in the D2 mice and only slightly in the D2B6 animals indicating a strong influence of the proximal region of Chr10 in the development of tolerance. Gene expression profiling and pattern matching identified 64, 53, 86, and 123 predisposition genes and 81, 96, 106, and 82 tolerance genes in the periaqueductal gray (PAG), prefrontal cortex, temporal lobe, and ventral striatum, respectively. A potential gene network was identified in the PAG in which 19 of the 34 genes were strongly associated with tolerance. Eleven of the network genes were found to reside in quantitative trait loci previously associated with morphine-related behaviors, whereas seven were predictive of tolerance (morphine-naive condition). Overall, the genes modified by chronic morphine administration show a strong presence in canonical pathways representative of neuroadaptation. A potentially significant role for the micro-RNA and epigenetic mechanisms in response to chronic administration of pharmacologically relevant doses of morphine was highlighted by candidate genes Dicer and H19.


Subject(s)
Analgesics, Opioid/pharmacology , Drug Tolerance/genetics , Gene Regulatory Networks/genetics , Morphine/pharmacology , Animals , Dose-Response Relationship, Drug , Female , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Genetics, Behavioral/methods , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred DBA , Pain Measurement/drug effects , Pain Measurement/methods
16.
Physiol Behav ; 86(3): 347-55, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16126238

ABSTRACT

Repeated administration of psychostimulants, such as amphetamine and cocaine, results in a long-lasting enhancement of behavioral responses elicited by a subsequent challenge injection of these drugs. This phenomenon has been termed behavioral sensitization. A well established model of individual differences based on the locomotor response to a novel environment has been shown to reliably predict the degree of behavioral sensitization to amphetamine. Rats that have high locomotor response in a novel environment (high responders or HR) develop greater behavioral sensitization to psychostimulants when compared to rats that show low locomotor activity in the same novel environment (low Responders or LR). Therefore, this model is ideal to study genetic factors that may underlie behavioral sensitization to psychostimulants. In this study, adult Sprague-Dawley rats were daily injected with amphetamine (1 mg/kg, i.p.) or saline for 9 days. Locomotor activity was recorded every other day. Following a one week-withdrawal a subsequent challenge of a lower dose of amphetamine (0.5 mg/kg, i.p.) was given to all rats (amphetamine pretreated and saline pretreated) and their locomotor activity was recorded. Our results show that HR rats, but not LR rats, develop behavioral sensitization to the locomotor activating effects of amphetamine. Furthermore, only HR rats pretreated with amphetamine exhibited an increase in dopamine transporter mRNA in the ventral tegmental area (VTA) and substantia nigra (SN). Tyrosine hydroxylase mRNA in the VTA and SN was upregulated in both HR and LR rats pretreated with amphetamine when compared to HR and LR rats pretreated with saline. These results demonstrate the existence of individual differences in behavioral sensitization to amphetamine and suggest that dopamine transporter, but not tyrosine hydroxylase, may be a critical factor in the development and expression of behavioral sensitization to the locomotor activating effects of amphetamine.


Subject(s)
Amphetamine/pharmacology , Behavior, Animal/drug effects , Dopamine Plasma Membrane Transport Proteins/physiology , Dopamine Uptake Inhibitors/pharmacology , Individuality , Analysis of Variance , Animals , Behavior, Animal/physiology , Cell Count/methods , Exploratory Behavior/drug effects , In Situ Hybridization/methods , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Statistics as Topic , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/physiology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...