Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Neuropsychopharmacology ; 45(2): 247-255, 2020 01.
Article in English | MEDLINE | ID: mdl-31005059

ABSTRACT

The medial habenula (MHb) is considered a brain center regulating aversive states. The mu opioid receptor (MOR) has been traditionally studied at the level of nociceptive and mesolimbic circuits, for key roles in pain relief and reward processing. MOR is also densely expressed in MHb, however, MOR function at this brain site is virtually unknown. Here we tested the hypothesis that MOR in the MHb (MHb-MOR) also regulates aversion processing. We used chnrb4-Cre driver mice to delete the Oprm1 gene in chnrb4-neurons, predominantly expressed in the MHb. Conditional mutant (B4MOR) mice showed habenula-specific reduction of MOR expression, restricted to chnrb4-neurons (50% MHb-MORs). We tested B4MOR mice in behavioral assays to evaluate effects of MOR activation by morphine, and MOR blockade by naloxone. Locomotor, analgesic, rewarding, and motivational effects of morphine were preserved in conditional mutants. In contrast, conditioned place aversion (CPA) elicited by naloxone was reduced in both naïve (high dose) and morphine-dependent (low dose) B4MOR mice. Further, physical signs of withdrawal precipitated by either MOR (naloxone) or nicotinic receptor (mecamylamine) blockade were attenuated. These data suggest that MORs expressed in MHb B4-neurons contribute to aversive effects of naloxone, including negative effect and aversive effects of opioid withdrawal. MORs are inhibitory receptors, therefore we propose that endogenous MOR signaling normally inhibits chnrb4-neurons of the MHb and moderates their known aversive activity, which is unmasked upon receptor blockade. Thus, in addition to facilitating reward at several brain sites, tonic MOR activity may also limit aversion within the MHb circuitry.


Subject(s)
Avoidance Learning/drug effects , Habenula/drug effects , Habenula/metabolism , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid, mu/deficiency , Animals , Avoidance Learning/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Opioid, mu/genetics
2.
Science ; 366(6462): 250-254, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31601771

ABSTRACT

The unconventional N-methyl-d-aspartate (NMDA) receptor subunits GluN3A and GluN3B can, when associated with the other glycine-binding subunit GluN1, generate excitatory conductances purely activated by glycine. However, functional GluN1/GluN3 receptors have not been identified in native adult tissues. We discovered that GluN1/GluN3A receptors are operational in neurons of the mouse adult medial habenula (MHb), an epithalamic area controlling aversive physiological states. In the absence of glycinergic neuronal specializations in the MHb, glial cells tuned neuronal activity via GluN1/GluN3A receptors. Reducing GluN1/GluN3A receptor levels in the MHb prevented place-aversion conditioning. Our study extends the physiological and behavioral implications of glycine by demonstrating its control of negatively valued emotional associations via excitatory glycinergic NMDA receptors.


Subject(s)
Behavior, Animal , Emotions , Glycine/metabolism , Habenula/metabolism , Nerve Tissue Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Calcium/metabolism , Cell Line , Conditioning, Psychological , Cues , Glycine/pharmacology , Humans , Mice , Mice, Knockout , Neuroglia/metabolism , Neurons/metabolism , Patch-Clamp Techniques
3.
eNeuro ; 6(4)2019.
Article in English | MEDLINE | ID: mdl-31346000

ABSTRACT

The orphan receptor GPR88 is highly expressed in D1 receptor (D1R)- and D2R-medium spiny neurons (MSNs) and has been associated to striatum-dependent functions in rodents. The total deletion of Gpr88 in mice was shown to decrease anxiety-like behaviors, increase stereotypies and locomotion, and impair motor coordination and motor learning. Knowing the opposing role of D1R- and D2R-MSNs, we here investigated the respective roles of GPR88 in the two MSN subtypes for these behaviors. To do so, we compared effects of a conditional Gpr88 gene knock-out (KO) in D1R-MSNs (D1R-Gpr88 mice) or D2R-MSNs (A2AR-Gpr88 mice) with effects of the total Gpr88 KO (CMV-Gpr88 mice). Overall, most phenotypes of CMV-Gpr88 mice were recapitulated in A2AR-Gpr88 mice, including reduced marble burying, increased social interactions, increased locomotor activity and stereotypies in the open field, and reduced motor coordination in the rotarod. Exceptions were the reduced habituation to the open field and reduced motor skill learning, which were observed in CMV-Gpr88 and D1R-Gpr88 mice, but not in A2AR-Gpr88 mice. D1R-Gpr88 mice otherwise showed no other phenotype in this study. Our data together show that GPR88 modulates the function of both D1R- and D2R-MSNs, and that GPR88 activity in these two neuron populations has very different and dissociable impacts on behavior. We suggest that GPR88 in D2R-MSNs shapes defensive and social behavior and contributes in maintaining the inhibition of basal ganglia outputs to control locomotion, stereotypies and motor coordination, while GPR88 in D1R-MSNs promotes novelty habituation and motor learning.


Subject(s)
Affect/physiology , Behavior, Animal/physiology , Corpus Striatum/physiology , Neurons/physiology , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Exploratory Behavior/physiology , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Skills/physiology , Social Behavior
4.
Genes Brain Behav ; 17(8): e12473, 2018 11.
Article in English | MEDLINE | ID: mdl-29575471

ABSTRACT

Mouse models are widely used to understand genetic bases of behavior. Traditional testing typically requires multiple experimental settings, captures only snapshots of behavior and involves human intervention. The recent development of automated home cage monitoring offers an alternative method to study mouse behavior in their familiar and social environment, and over weeks. Here, we used the IntelliCage system to test this approach for mouse phenotyping, and studied mice lacking Gpr88 that have been extensively studied using standard testing. We monitored mouse behavior over 22 days in 4 different phases. In the free adaptation phase, Gpr88 -/- mice showed delayed habituation to the home cage, and increased frequency of same corner returns behavior in their alternation pattern. In the following nose-poke adaptation phase, non-habituation continued, however, mutant mice acquired nose-poke conditioning similar to controls. In the place learning and reversal phase, Gpr88-/- mice developed preference for the water/sucrose corner with some delay, but did not differ from controls for reversal. Finally, in a fixed schedule-drinking phase, control animals showed higher activity during the hour preceding water accessibility, and reduced activity after access to water was terminated. Mutant mice did not show this behavior, showing lack of anticipatory behavior. Our data therefore confirm hyperactivity, non-habituation and altered exploratory behaviors that were reported previously. Learning deficits described in other settings were barely detectable, and a novel phenotype was discovered. Home cage monitoring therefore extends previous findings and shows yet another facet of GPR88 function that deserves further investigation.


Subject(s)
Behavior Observation Techniques/instrumentation , Exploratory Behavior/physiology , Receptors, G-Protein-Coupled/metabolism , Animals , Behavior Observation Techniques/methods , Behavior, Animal/physiology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Motor Activity/physiology , Phenotype , Receptors, G-Protein-Coupled/genetics
5.
Mol Psychiatry ; 23(10): 2050-2056, 2018 10.
Article in English | MEDLINE | ID: mdl-29158579

ABSTRACT

Bipolar disorder (BD) is a prevalent mood disorder that tends to cluster in families. Despite high heritability estimates, few genetic susceptibility factors have been identified over decades of genetic research. One possible interpretation for the shortcomings of previous studies to detect causative genes is that BD is caused by highly penetrant rare variants in many genes. We explored this hypothesis by sequencing the exomes of affected individuals from 40 well-characterized multiplex families. We identified rare variants segregating with affected status in many interesting genes, and found an enrichment of deleterious variants in G protein-coupled receptor (GPCR) family genes, which are important drug targets. Furthermore, we showed targeted downstream GPCR dysregulation for some of the variants that may contribute to disease pathology. Particularly interesting was the finding of a rare and functionally relevant nonsense mutation in the corticotropin-releasing hormone receptor 2 (CRHR2) gene that tracked with affected status in one family. By focusing on rare variants in informative families, we identified key biochemical pathways likely implicated in this complex disorder.


Subject(s)
Bipolar Disorder/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Adult , Bipolar Disorder/metabolism , Case-Control Studies , Family , Female , Gene Frequency/genetics , Genetic Linkage , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Middle Aged , Pedigree , Receptors, Corticotropin-Releasing Hormone/genetics , Exome Sequencing
6.
Mol Psychiatry ; 20(10): 1219-31, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25330738

ABSTRACT

MicroRNAs (miRNAs) induce messenger RNA (mRNA) degradation and repress mRNA translation. Several miRNAs control the expression of the brain-derived neurotrophic factor (BDNF) in the prefrontal cortex (PFC). The BDNF signaling pathway is activated by moderate intake of alcohol to prevent escalation to excessive drinking. Here, we present data to suggest that the transition from moderate to uncontrolled alcohol intake occurs, in part, upon a breakdown of this endogenous protective pathway via a miRNA-dependent mechanism. Specifically, a mouse paradigm that mimics binge alcohol drinking in humans produced a robust reduction in BDNF mRNA levels in the medial PFC (mPFC), which was associated with increased expression of several miRNAs including miR-30a-5p. We show that miR-30a-5p binds the 3' untranslated region of BDNF, and that overexpression of miR-30a-5p in the mPFC decreased BDNF expression. Importantly, overexpression of miR-30a-5p in the mPFC produced an escalation of alcohol intake and a preference over water. Conversely, inhibition of miR-30a-5p in the mPFC using a Locked Nucleic Acid sequence that targets miR-30a-5p restored BDNF levels and decreased excessive alcohol intake. Together, our results indicate that miR-30a-5p plays a key role in the transition from moderate to excessive alcohol intake.


Subject(s)
Alcohol Drinking/genetics , Alcoholic Intoxication/genetics , MicroRNAs/genetics , Prefrontal Cortex/physiology , Alcohol Drinking/metabolism , Alcoholic Intoxication/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Prefrontal Cortex/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Transl Psychiatry ; 3: e231, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23423140

ABSTRACT

Harmful excessive use of alcohol has a severe impact on society and it remains one of the major causes of morbidity and mortality in the population. However, mechanisms that underlie excessive alcohol consumption are still poorly understood, and thus available medications for alcohol use disorders are limited. Here, we report that changing the level of chromatin condensation by affecting DNA methylation or histone acetylation limits excessive alcohol drinking and seeking behaviors in rodents. Specifically, we show that decreasing DNA methylation by inhibiting the activity of DNA methyltransferase (DNMT) with systemic administration of the FDA-approved drug, 5-azacitidine (5-AzaC) prevents excessive alcohol use in mice. Similarly, we find that increasing histone acetylation via systemic treatment with several histone deacetylase (HDAC) inhibitors reduces mice binge-like alcohol drinking. We further report that systemic administration of the FDA-approved HDAC inhibitor, SAHA, inhibits the motivation of rats to seek alcohol. Importantly, the actions of both DNMT and HDAC inhibitors are specific for alcohol, as no changes in saccharin or sucrose intake were observed. In line with these behavioral findings, we demonstrate that excessive alcohol drinking increases DNMT1 levels and reduces histone H4 acetylation in the nucleus accumbens (NAc) of rodents. Together, our findings illustrate that DNA methylation and histone acetylation control the level of excessive alcohol drinking and seeking behaviors in preclinical rodent models. Our study therefore highlights the possibility that DNMT and HDAC inhibitors can be used to treat harmful alcohol abuse.


Subject(s)
Alcoholism/drug therapy , Alcoholism/metabolism , Behavior, Animal/drug effects , Chromatin Assembly and Disassembly/drug effects , Chromatin Assembly and Disassembly/genetics , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Histone Deacetylase Inhibitors/administration & dosage , Acetylation/drug effects , Alcoholism/enzymology , Animals , Azacitidine/administration & dosage , Azacitidine/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Disease Models, Animal , Histones/drug effects , Humans , Mice , Rats
9.
Eur J Neurosci ; 27(11): 2973-84, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18588537

ABSTRACT

Addiction develops from the gradual adaptation of the brain to chronic drug exposure, and involves genetic reprogramming of neuronal function. The central extended amygdala (EAc) is a network formed by the central amygdala and the bed nucleus of the stria terminalis. This key site controls drug craving and seeking behaviors, and has not been investigated at the gene regulation level. We used Affymetrix microarrays to analyze transcriptional activity in the murine EAc, with a focus on mu-opioid receptor-associated events because these receptors mediate drug reward and dependence. We identified 132 genes whose expression is regulated by a chronic escalating morphine regimen in the EAc from wild-type but not mu-opioid receptor knockout mice. These modifications are mostly EAc-specific. Gene ontology analysis reveals an overrepresentation of neurogenesis, cell growth and signaling protein categories. A separate quantitative PCR analysis of genes in the last of these groups confirms the dysregulation of both orphan (Gpr88) and known (DrD1A, Adora2A, Cnr1, Grm5, Gpr6) G protein-coupled receptors, scaffolding (PSD95, Homer1) and signaling (Sgk, Cap1) proteins, and neuropeptides (CCK, galanin). These transcriptional modifications do not occur following a single morphine injection, and hence result from long-term adaptation to excessive mu receptor activation. Proteins encoded by these genes are classically associated with spine modules function in other brain areas, and therefore our data suggest a remodeling of EAc circuits at sites where glutamatergic and monoaminergic afferences interact. Together, mu receptor-dependent genes identified in this study potentially contribute to drug-induced neural plasticity, and provide a unique molecular repertoire towards understanding drug craving and relapse.


Subject(s)
Amygdala/drug effects , Analgesics, Opioid/pharmacology , Gene Expression Regulation/drug effects , Neuronal Plasticity/drug effects , Receptors, Opioid, mu/agonists , Transcriptional Activation/drug effects , Amygdala/metabolism , Animals , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/pharmacology , Neural Pathways/drug effects , Neural Pathways/metabolism , Neuronal Plasticity/genetics , Oligonucleotide Array Sequence Analysis , Opioid-Related Disorders/genetics , Opioid-Related Disorders/metabolism , Opioid-Related Disorders/physiopathology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Opioid, mu/metabolism , Septal Nuclei/drug effects , Septal Nuclei/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptional Activation/genetics
10.
Ann N Y Acad Sci ; 1129: 175-84, 2008.
Article in English | MEDLINE | ID: mdl-18591478

ABSTRACT

The lateral hypothalamus (LH) is a brain structure that controls hedonic properties of both natural rewards and drugs of abuse. Mu opioid receptors are known to mediate drug reward, but whether overstimulation of these receptors impacts on LH function has not been studied. Here we have used a genome-wide microarray approach to identify LH responses to chronic mu opioid receptor activation at the transcriptional level. We have subjected wild-type and mu opioid receptor knockout mice to an escalating morphine regimen, which produces severe physical dependence in wild-type but not mutant animals. We have analyzed gene profiles in LH samples using the 430A.2 Affymetrix array and identified a set of 25 genes whose expression is altered by morphine in wild-type mice only. The regulation was confirmed for a subset of these genes using real-time quantitative PCR on samples from independent treatments. Altered expression of aquaporin 4, apolipoprotein D, and prostaglandin synthase is indicative of modified LH physiology. The regulation of two signaling genes (the serum glucocorticoid kinase and the regulator of G protein signaling 4) suggests that neurotransmission is altered in LH circuitry. Finally, the downregulation of apelin may indicate a potential role for this neuropeptide in opioid signaling and hedonic homeostasis. Altogether, our study shows that chronic mu opioid receptor stimulation induces gene expression plasticity in the LH and provides a unique collection of mu opioid receptor-dependent genes that potentially contribute to alter reward processes in addictive diseases.


Subject(s)
Gene Expression Regulation , Hypothalamus/metabolism , Receptors, Opioid, mu/genetics , Animals , Gene Expression Regulation/drug effects , Hypothalamus/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/pharmacology , Polymerase Chain Reaction , Receptors, Opioid, mu/metabolism , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...