Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Biol Psychiatry ; 95(2): 123-135, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-37207936

ABSTRACT

BACKGROUND: Deficient social interactions are a hallmark of major neuropsychiatric disorders, and accumulating evidence points to altered social reward and motivation as key underlying mechanisms of these pathologies. In the present study, we further explored the role of the balance of activity between D1 and D2 receptor-expressing striatal projection neurons (D1R- and D2R-SPNs) in the control of social behavior, challenging the hypothesis that excessive D2R-SPN activity, rather than deficient D1R-SPN activity, compromises social behavior. METHODS: We selectively ablated D1R- and D2R-SPNs using an inducible diphtheria toxin receptor-mediated cell targeting strategy and assessed social behavior as well as repetitive/perseverative behavior, motor function, and anxiety levels. We tested the effects of optogenetic stimulation of D2R-SPNs in the nucleus accumbens (NAc) and pharmacological compounds repressing D2R-SPN. RESULTS: Targeted deletion of D1R-SPNs in the NAc blunted social behavior in mice, facilitated motor skill learning, and increased anxiety levels. These behaviors were normalized by pharmacological inhibition of D2R-SPN, which also repressed transcription in the efferent nucleus, the ventral pallidum. Ablation of D1R-SPNs in the dorsal striatum had no impact on social behavior but impaired motor skill learning and decreased anxiety levels. Deletion of D2R-SPNs in the NAc produced motor stereotypies but facilitated social behavior and impaired motor skill learning. We mimicked excessive D2R-SPN activity by optically stimulating D2R-SPNs in the NAc and observed a severe deficit in social interaction that was prevented by D2R-SPN pharmacological inhibition. CONCLUSIONS: Repressing D2R-SPN activity may represent a promising therapeutic strategy to relieve social deficits in neuropsychiatric disorders.


Subject(s)
Neurons , Nucleus Accumbens , Mice , Animals , Neurons/physiology , Social Behavior , Motivation , Learning , Receptors, Dopamine D1/metabolism
2.
Nat Chem Biol ; 18(8): 894-903, 2022 08.
Article in English | MEDLINE | ID: mdl-35681029

ABSTRACT

Membrane proteins, including ion channels, receptors and transporters, are often composed of multiple subunits and can form large complexes. Their specific composition in native tissues is difficult to determine and remains largely unknown. In this study, we developed a method for determining the subunit composition of endogenous cell surface protein complexes from isolated native tissues. Our method relies on nanobody-based sensors, which enable proximity detection between subunits in time-resolved Förster resonance energy transfer (FRET) measurements. Additionally, given conformation-specific nanobodies, the activation of these complexes can be recorded in native brain tissue. Applied to the metabotropic glutamate receptors in different brain regions, this approach revealed the clear existence of functional metabotropic glutamate (mGlu)2-mGlu4 heterodimers in addition to mGlu2 and mGlu4 homodimers. Strikingly, the mGlu4 subunits appear to be mainly heterodimers in the brain. Overall, these versatile biosensors can determine the presence and activity of endogenous membrane proteins in native tissues with high fidelity and convenience.


Subject(s)
Glutamic Acid , Receptors, Metabotropic Glutamate , Brain/metabolism , Fluorescence Resonance Energy Transfer/methods , Receptors, Metabotropic Glutamate/metabolism
3.
Neuropsychopharmacology ; 47(9): 1680-1692, 2022 08.
Article in English | MEDLINE | ID: mdl-35418620

ABSTRACT

Autism Spectrum Disorders (ASD) are neurodevelopmental disorders whose diagnosis relies on deficient social interaction and communication together with repetitive behavior. To date, no pharmacological treatment has been approved that ameliorates social behavior in patients with ASD. Based on the excitation/inhibition imbalance theory of autism, we hypothesized that bromide ions, long used as an antiepileptic medication, could relieve core symptoms of ASD. We evaluated the effects of chronic sodium bromide (NaBr) administration on autistic-like symptoms in three genetic mouse models of autism: Oprm1-/-, Fmr1-/- and Shank3Δex13-16-/- mice. We showed that chronic NaBr treatment relieved autistic-like behaviors in these three models. In Oprm1-/- mice, these beneficial effects were superior to those of chronic bumetanide administration. At transcriptional level, chronic NaBr in Oprm1 null mice was associated with increased expression of genes coding for chloride ions transporters, GABAA receptor subunits, oxytocin and mGlu4 receptor. Lastly, we uncovered synergistic alleviating effects of chronic NaBr and a positive allosteric modulator (PAM) of mGlu4 receptor on autistic-like behavior in Oprm1-/- mice. We evidenced in heterologous cells that bromide ions behave as PAMs of mGlu4, providing a molecular mechanism for such synergy. Our data reveal the therapeutic potential of bromide ions, alone or in combination with a PAM of mGlu4 receptor, for the treatment of ASDs.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Animals , Autism Spectrum Disorder/drug therapy , Autistic Disorder/drug therapy , Behavior, Animal , Bromides/pharmacology , Bromides/therapeutic use , Disease Models, Animal , Fragile X Mental Retardation Protein , Mice , Mice, Knockout , Microfilament Proteins/pharmacology , Microfilament Proteins/therapeutic use , Nerve Tissue Proteins/genetics , Receptors, GABA-A , Social Behavior , Sodium Compounds
4.
Microbiome ; 9(1): 157, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34238386

ABSTRACT

BACKGROUND: Autism spectrum disorders (ASD) are associated with dysregulation of the microbiota-gut-brain axis, changes in microbiota composition as well as in the fecal, serum, and urine levels of microbial metabolites. Yet a causal relationship between dysregulation of the microbiota-gut-brain axis and ASD remains to be demonstrated. Here, we hypothesized that the microbial metabolite p-Cresol, which is more abundant in ASD patients compared to neurotypical individuals, could induce ASD-like behavior in mice. RESULTS: Mice exposed to p-Cresol for 4 weeks in drinking water presented social behavior deficits, stereotypies, and perseverative behaviors, but no changes in anxiety, locomotion, or cognition. Abnormal social behavior induced by p-Cresol was associated with decreased activity of central dopamine neurons involved in the social reward circuit. Further, p-Cresol induced changes in microbiota composition and social behavior deficits could be transferred from p-Cresol-treated mice to control mice by fecal microbiota transplantation (FMT). We also showed that mice transplanted with the microbiota of p-Cresol-treated mice exhibited increased fecal p-Cresol excretion, compared to mice transplanted with the microbiota of control mice. In addition, we identified possible p-Cresol bacterial producers. Lastly, the microbiota of control mice rescued social interactions, dopamine neurons excitability, and fecal p-Cresol levels when transplanted to p-Cresol-treated mice. CONCLUSIONS: The microbial metabolite p-Cresol induces selectively ASD core behavioral symptoms in mice. Social behavior deficits induced by p-Cresol are dependant on changes in microbiota composition. Our study paves the way for therapeutic interventions targeting the microbiota and p-Cresol production to treat patients with ASD. Video abstract.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Gastrointestinal Microbiome , Animals , Autistic Disorder/etiology , Cresols , Fecal Microbiota Transplantation , Humans , Mice
5.
Neuropsychopharmacology ; 46(7): 1373-1385, 2021 06.
Article in English | MEDLINE | ID: mdl-33349673

ABSTRACT

Understanding the neurobiological underpinnings of abstinence from drugs of abuse is critical to allow better recovery and ensure relapse prevention in addicted subjects. By comparing the long-term transcriptional consequences of morphine and cocaine exposure, we identified the metabotropic glutamate receptor subtype 4 (mGluR4) as a promising pharmacological target in morphine abstinence. We evaluated the behavioral and molecular effects of facilitating mGluR4 activity in abstinent mice. Transcriptional regulation of marker genes of medium spiny neurons (MSNs) allowed best discriminating between 4-week morphine and cocaine abstinence in the nucleus accumbens (NAc). Among these markers, Grm4, encoding mGluR4, displayed down-regulated expression in the caudate putamen and NAc of morphine, but not cocaine, abstinent mice. Chronic administration of the mGluR4 positive allosteric modulator (PAM) VU0155041 (2.5 and 5 mg/kg) rescued social behavior, normalized stereotypies and anxiety and blunted locomotor sensitization in morphine abstinent mice. This treatment improved social preference but increased stereotypies in cocaine abstinent mice. Finally, the beneficial behavioral effects of VU0155041 treatment in morphine abstinent mice were correlated with restored expression of key MSN and neural activity marker genes in the NAc. This study reports that chronic administration of the mGluR4 PAM VU0155041 relieves long-term deleterious consequences of morphine exposure. It illustrates the neurobiological differences between opiate and psychostimulant abstinence and points to pharmacological repression of excessive activity of D2-MSNs in the NAc as a promising therapeutic lever in drug addiction.


Subject(s)
Cocaine , Receptors, Metabotropic Glutamate , Animals , Male , Mice , Mice, Inbred C57BL , Morphine/pharmacology , Nucleus Accumbens
6.
Mol Metab ; 21: 22-35, 2019 03.
Article in English | MEDLINE | ID: mdl-30686771

ABSTRACT

OBJECTIVES: The Fragile X Mental Retardation Protein (FMRP) is a widely expressed RNA-binding protein involved in translation regulation. Since the absence of FMRP leads to Fragile X Syndrome (FXS) and autism, FMRP has been extensively studied in brain. The functions of FMRP in peripheral organs and on metabolic homeostasis remain elusive; therefore, we sought to investigate the systemic consequences of its absence. METHODS: Using metabolomics, in vivo metabolic phenotyping of the Fmr1-KO FXS mouse model and in vitro approaches, we show that the absence of FMRP induced a metabolic shift towards enhanced glucose tolerance and insulin sensitivity, reduced adiposity, and increased ß-adrenergic-driven lipolysis and lipid utilization. RESULTS: Combining proteomics and cellular assays, we highlight that FMRP loss increased hepatic protein synthesis and impacted pathways notably linked to lipid metabolism. Mapping metabolomic and proteomic phenotypes onto a signaling and metabolic network, we predicted that the coordinated metabolic response to FMRP loss was mediated by dysregulation in the abundances of specific hepatic proteins. We experimentally validated these predictions, demonstrating that the translational regulator FMRP associates with a subset of mRNAs involved in lipid metabolism. Finally, we highlight that FXS patients mirror metabolic variations observed in Fmr1-KO mice with reduced circulating glucose and insulin and increased free fatty acids. CONCLUSIONS: Loss of FMRP results in a widespread coordinated systemic response that notably involves upregulation of protein translation in the liver, increased utilization of lipids, and significant changes in metabolic homeostasis. Our study unravels metabolic phenotypes in FXS and further supports the importance of translational regulation in the homeostatic control of systemic metabolism.


Subject(s)
Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Glucose/metabolism , Lipolysis , Adipocytes/metabolism , Animals , Disease Models, Animal , Fatty Acids, Nonesterified/metabolism , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/blood , Fragile X Syndrome/pathology , Gene Knockout Techniques , Glucose/analysis , Homeostasis , Humans , Insulin/analysis , Insulin/metabolism , Leptin/metabolism , Liver/metabolism , Male , Metabolomics , Mice , Mice, Knockout , Protein Biosynthesis , Proteomics , RNA, Messenger/metabolism
7.
Transl Psychiatry ; 8(1): 197, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30242222

ABSTRACT

The mu opioid receptor (MOR) plays a critical role in modulating social behavior in humans and animals. Accordingly, MOR null mice display severe alterations in their social repertoire as well as multiple other behavioral deficits, recapitulating core and secondary symptoms of autism spectrum disorder (ASD). Such behavioral profile suggests that MOR dysfunction, and beyond this, altered reward processes may contribute to ASD etiopathology. Interestingly, the only treatments that proved efficacy in relieving core symptoms of ASD, early behavioral intervention programs, rely principally on positive reinforcement to ameliorate behavior. The neurobiological underpinnings of their beneficial effects, however, remain poorly understood. Here we back-translated applied behavior analysis (ABA)-based behavioral interventions to mice lacking the MOR (Oprm1-/-), as a model of autism with blunted reward processing. By associating a positive reinforcement, palatable food reward, to daily encounter with a wild-type congener, we were able to rescue durably social interaction and preference in Oprm1-/- mice. Along with behavioral improvements, the expression of marker genes of neuronal activity and plasticity as well as genes of the oxytocin/vasopressin system were remarkably normalized in the reward/social circuitry. Our study provides further evidence for a critical involvement of reward processes in driving social behavior and opens new perspectives regarding therapeutic intervention in ASD.


Subject(s)
Autism Spectrum Disorder/therapy , Behavior Therapy , Behavior, Animal , Receptors, Opioid, mu/genetics , Reward , Social Behavior , Animals , Applied Behavior Analysis , Autism Spectrum Disorder/genetics , Disease Models, Animal , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/genetics
8.
J Med Chem ; 61(19): 8670-8692, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30199637

ABSTRACT

Oxytocin (OT) and its receptor (OT-R) are implicated in the etiology of autism spectrum disorders (ASD), and OT-R is a potential target for therapeutic intervention. Very few nonpeptide oxytocin agonists have currently been reported. Their molecular and in vivo pharmacology remain to be clarified, and none of them has been shown to be efficient in improving social interaction in animal models relevant to ASD. In an attempt to rationalize the design of centrally active nonpeptide full agonists, we studied in a systematic way the structural determinants of the affinity and efficacy of representative ligands of the V1a and V2 vasopressin receptor subtypes (V1a-R and V2-R) and of the oxytocin receptor. Our results confirm the subtlety of the structure-affinity and structure-efficacy relationships around vasopressin/oxytocin receptor ligands and lead however to the first nonpeptide OT receptor agonist active in a mouse model of ASD after peripheral ip administration.


Subject(s)
Autistic Disorder/drug therapy , Disease Models, Animal , Interpersonal Relations , Psychotropic Drugs/pharmacology , Pyrazoles/pharmacology , Pyrrolidines/pharmacology , Receptors, Opioid, mu/physiology , Receptors, Oxytocin/administration & dosage , Receptors, Oxytocin/agonists , Animals , Autistic Disorder/psychology , Blood-Brain Barrier/drug effects , Female , HEK293 Cells , Humans , Ligands , Male , Mice , Mice, Knockout , Psychotropic Drugs/chemistry , Pyrazoles/therapeutic use , Pyrrolidines/therapeutic use , Receptors, Oxytocin/therapeutic use , Structure-Activity Relationship
9.
Br J Pharmacol ; 175(14): 2750-2769, 2018 07.
Article in English | MEDLINE | ID: mdl-28369738

ABSTRACT

The endogenous opioid system is well known to relieve pain and underpin the rewarding properties of most drugs of abuse. Among opioid receptors, the µ receptor mediates most of the analgesic and rewarding properties of opioids. Based on striking similarities between social distress, physical pain and opiate withdrawal, µ receptors have been proposed to play a critical role in modulating social behaviour in humans and animals. This review summarizes experimental data demonstrating such role and proposes a novel model, the µ opioid receptor balance model, to account for the contribution of µ receptors to the subtle regulation of social behaviour. Interestingly, µ receptor null mice show behavioural deficits similar to those observed in patients with autism spectrum disorder (ASD), including severe impairment in social interactions. Therefore, after a brief summary of recent evidence for blunted (social) reward processes in subjects with ASD, we review here arguments for altered µ receptor function in this pathology. This article is part of a themed section on Emerging Areas of Opioid Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.14/issuetoc.


Subject(s)
Autism Spectrum Disorder/metabolism , Receptors, Opioid, mu/metabolism , Reward , Social Behavior , Animals , Humans , Receptors, Opioid, mu/genetics
10.
Addict Biol ; 22(5): 1205-1217, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27126842

ABSTRACT

Unified theories of addiction are challenged by differing drug-seeking behaviors and neurobiological adaptations across drug classes, particularly for narcotics and psychostimulants. We previously showed that protracted abstinence to opiates leads to despair behavior and social withdrawal in mice, and we identified a transcriptional signature in the extended amygdala that was also present in animals abstinent from nicotine, Δ9-tetrahydrocannabinol (THC) and alcohol. Here we examined whether protracted abstinence to these four drugs would also share common behavioral features, and eventually differ from abstinence to the prototypic psychostimulant cocaine. We found similar reduced social recognition, increased motor stereotypies and increased anxiety with relevant c-fos response alterations in morphine, nicotine, THC and alcohol abstinent mice. Protracted abstinence to cocaine, however, led to strikingly distinct, mostly opposing adaptations at all levels, including behavioral responses, neuronal activation and gene expression. Together, these data further document the existence of common hallmarks for protracted abstinence to opiates, nicotine, THC and alcohol that develop within motivation/emotion brain circuits. In our model, however, these do not apply to cocaine, supporting the notion of unique mechanisms in psychostimulant abuse.


Subject(s)
Amygdala/metabolism , Analgesics, Opioid/adverse effects , Behavior, Animal , Cannabinoid Receptor Agonists/adverse effects , Central Nervous System Depressants/adverse effects , Dopamine Uptake Inhibitors/adverse effects , Nicotinic Agonists/adverse effects , Social Behavior , Substance Withdrawal Syndrome/physiopathology , Alcohol Abstinence , Animals , Anxiety/psychology , Brain/metabolism , Cocaine/adverse effects , Dronabinol/adverse effects , Drug-Seeking Behavior , Emotions , Ethanol/adverse effects , Male , Mice , Morphine/adverse effects , Motivation , Nicotine/adverse effects , Proto-Oncogene Proteins c-fos/metabolism , Stereotyped Behavior/physiology , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology
11.
J Stud Alcohol Drugs ; 77(5): 692-5, 2016 09.
Article in English | MEDLINE | ID: mdl-27588526

ABSTRACT

Quitting drug abuse represents a true challenge for addicted individuals because of the highly persistent vulnerability to relapse. Identifying long-lasting, drug-induced alterations in the brain-including at the transcriptome level-that underlie such vulnerability appears invaluable to improve relapse prevention. Despite substantial technological developments and research effort, however, the picture of drug-induced adaptations provided by high-throughput transcriptomics remains frustratingly partial, notably because of methodological issues. Major advances were made, however, regarding the time course and specificity of long-term transcriptional consequences of drug exposure as well as the recruitment of small, noncoding mRNAs (or miRNAs [microRNAs]) that were previously undetectable. Most importantly, high-throughput studies have benefited from systems biology approaches and shifted their interest toward regulations within functional gene networks rather than individual changes. Such network-based gene discovery approaches have proven informative to delineate the physiological processes, cellular signaling pathways, and neuronal populations altered by drug exposure. Provided the high-throughput effort will be pursued, together with the development of adapted bioinformatics tools, addiction transcriptomics should progressively integrate data across multiple scales (from epigenome to protein), allowing a better understanding of the genetics of drug abuse and opening novel therapeutic trails.


Subject(s)
Behavior, Addictive/genetics , Comprehension , Cues , Substance-Related Disorders/genetics , Transcription, Genetic/genetics , Adaptation, Physiological/genetics , Behavior, Addictive/diagnosis , Behavior, Addictive/therapy , Brain/physiology , Computational Biology/trends , Humans , Recurrence , Substance-Related Disorders/diagnosis , Substance-Related Disorders/therapy , Time Factors
12.
Proc Natl Acad Sci U S A ; 113(26): E3619-28, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27233938

ABSTRACT

Fragile X syndrome (FXS) is caused by the absence of the Fragile X Mental Retardation Protein (FMRP) in neurons. In the mouse, the lack of FMRP is associated with an excessive translation of hundreds of neuronal proteins, notably including postsynaptic proteins. This local protein synthesis deregulation is proposed to underlie the observed defects of glutamatergic synapse maturation and function and to affect preferentially the hundreds of mRNA species that were reported to bind to FMRP. How FMRP impacts synaptic protein translation and which mRNAs are most important for the pathology remain unclear. Here we show by cross-linking immunoprecipitation in cortical neurons that FMRP is mostly associated with one unique mRNA: diacylglycerol kinase kappa (Dgkκ), a master regulator that controls the switch between diacylglycerol and phosphatidic acid signaling pathways. The absence of FMRP in neurons abolishes group 1 metabotropic glutamate receptor-dependent DGK activity combined with a loss of Dgkκ expression. The reduction of Dgkκ in neurons is sufficient to cause dendritic spine abnormalities, synaptic plasticity alterations, and behavior disorders similar to those observed in the FXS mouse model. Overexpression of Dgkκ in neurons is able to rescue the dendritic spine defects of the Fragile X Mental Retardation 1 gene KO neurons. Together, these data suggest that Dgkκ deregulation contributes to FXS pathology and support a model where FMRP, by controlling the translation of Dgkκ, indirectly controls synaptic proteins translation and membrane properties by impacting lipid signaling in dendritic spine.


Subject(s)
Diacylglycerol Kinase/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Neurons/enzymology , Aged , Animals , Dendritic Spines/enzymology , Dendritic Spines/metabolism , Diacylglycerol Kinase/genetics , Diglycerides/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/enzymology , Fragile X Syndrome/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neurons/metabolism , Signal Transduction
13.
J Comp Neurol ; 524(14): 2776-802, 2016 10 01.
Article in English | MEDLINE | ID: mdl-26918661

ABSTRACT

GPR88 is a neuronal cerebral orphan G-protein-coupled receptor (GPCR) that has been linked to various psychiatric disorders. However, no extensive description of its localization has been provided so far. Here, we investigate the spatiotemporal expression of the GPR88 in prenatal and postnatal rat tissues by using in situ hybridization and immunohistochemistry. GPR88 protein was initially detected at embryonic day 16 (E16) in the striatal primordium. From E16-E20 to adulthood, the highest expression levels of both protein and mRNA were observed in striatum, olfactory tubercle, nucleus accumbens, amygdala, and neocortex, whereas in spinal cord, pons, and medulla GPR88 expression remains discrete. We observed an intracellular redistribution of GPR88 during cortical lamination. In the cortical plate of the developing cortex, GPR88 presents a classical GPCR plasma membrane/cytoplasmic localization that shifts, on the day of birth, to nuclei of neurons progressively settling in layers V to II. This intranuclear localization remains throughout adulthood and was also detected in monkey and human cortex as well as in the amygdala and hypothalamus of rats. Apart from the central nervous system, GPR88 was transiently expressed at high levels in peripheral tissues, including adrenal cortex (E16-E21) and cochlear ganglia (E19-P3), and also at moderate levels in retina (E18-E19) and spleen (E21-P7). The description of the GPR88 anatomical expression pattern may provide precious functional insights into this novel receptor. Furthermore, the GRP88 nuclear localization suggests nonclassical GPCR modes of action of the protein that could be relevant for cortical development and psychiatric disorders. J. Comp. Neurol. 524:2776-2802, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Nucleus/metabolism , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Cytoplasm/metabolism , Gene Expression Regulation, Developmental , Receptors, G-Protein-Coupled/biosynthesis , Age Factors , Animals , Animals, Newborn , Cerebral Cortex/chemistry , Cytoplasm/chemistry , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/analysis , Young Adult
14.
Biol Psychiatry ; 79(11): 917-27, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26188600

ABSTRACT

BACKGROUND: GPR88 is an orphan G protein coupled receptor highly enriched in the striatum, and previous studies have focused on GPR88 function in striatal physiology. The receptor is also expressed in other brain areas, and here we examined whether GPR88 function extends beyond striatal-mediated responses. METHODS: We created Gpr88 knockout mice and examined both striatal and extrastriatal regions at molecular and cellular levels. We also tested striatum-, hippocampus-, and amygdala-dependent behaviors in Gpr88(-/-) mice using extensive behavioral testing. RESULTS: We found increased G protein coupling for delta opioid receptor (DOR) and mu opioid, but not other Gi/o coupled receptors, in the striatum of Gpr88 knockout mice. We also found modifications in gene transcription, dopamine and serotonin contents, and dendritic morphology inside and outside the striatum. Behavioral testing confirmed striatal deficits (hyperactivity, stereotypies, motor impairment in rotarod). In addition, mutant mice performed better in spatial tasks dependent on hippocampus (Y-maze, novel object recognition, dual solution cross-maze) and also showed markedly reduced levels of anxiety (elevated plus maze, marble burying, novelty suppressed feeding). Strikingly, chronic blockade of DOR using naltrindole partially improved motor coordination and normalized spatial navigation and anxiety of Gpr88(-/-) mice. CONCLUSIONS: We demonstrate that GPR88 is implicated in a large repertoire of behavioral responses that engage motor activity, spatial learning, and emotional processing. Our data also reveal functional antagonism between GPR88 and DOR activities in vivo. The therapeutic potential of GPR88 therefore extends to cognitive and anxiety disorders, possibly in interaction with other receptor systems.


Subject(s)
Anxiety/metabolism , Movement Disorders/metabolism , Receptors, G-Protein-Coupled/deficiency , Spatial Learning/physiology , Amygdala/metabolism , Amygdala/pathology , Animals , Anxiety/drug therapy , Anxiety/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dendrites/drug effects , Dendrites/metabolism , Dendrites/pathology , Dopamine/metabolism , Female , Hippocampus/metabolism , Hippocampus/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Movement Disorders/drug therapy , Movement Disorders/pathology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, G-Protein-Coupled/genetics , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Serotonin/metabolism , Spatial Learning/drug effects
15.
Neuropsychopharmacology ; 39(9): 2049-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24619243

ABSTRACT

The etiology of Autism Spectrum Disorders (ASDs) remains largely unknown. Identifying vulnerability genes for autism represents a major challenge in the field and allows the development of animal models for translational research. Mice lacking the mu opioid receptor gene (Oprm1(-/-)) were recently proposed as a monogenic mouse model of autism, based on severe deficits in social behavior and communication skills. We confirm this hypothesis by showing that adult Oprm1(-/-) animals recapitulate core and multiple comorbid behavioral symptoms of autism and also display anatomical, neurochemical, and genetic landmarks of the disease. Chronic facilitation of mGluR4 signaling, which we identified as a novel pharmacological target in ASDs in these mice, was more efficient in alleviating behavioral deficits than the reference molecule risperidone. Altogether, our data provide first evidence that disrupted mu opioid receptor signaling is sufficient to trigger a comprehensive autistic syndrome, maybe through blunted social reward processes, and this mouse model opens promising avenues for therapeutic innovation.


Subject(s)
Child Development Disorders, Pervasive/pathology , Child Development Disorders, Pervasive/physiopathology , Receptors, Metabotropic Glutamate/metabolism , Receptors, Opioid, mu/metabolism , Aggression/drug effects , Aggression/physiology , Anilides/pharmacology , Animals , Anxiety/drug therapy , Anxiety/pathology , Anxiety/physiopathology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/pathology , Brain/physiopathology , Child Development Disorders, Pervasive/drug therapy , Convulsants/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Disease Models, Animal , Excitatory Amino Acid Agents/pharmacology , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Pentylenetetrazole/pharmacology , Receptors, Opioid, mu/genetics , Seizures/chemically induced , Seizures/physiopathology , Social Behavior
16.
PLoS One ; 8(9): e74706, 2013.
Article in English | MEDLINE | ID: mdl-24069332

ABSTRACT

Opiates are powerful drugs to treat severe pain, and act via mu opioid receptors distributed throughout the nervous system. Their clinical use is hampered by centrally-mediated adverse effects, including nausea or respiratory depression. Here we used a genetic approach to investigate the potential of peripheral mu opioid receptors as targets for pain treatment. We generated conditional knockout (cKO) mice in which mu opioid receptors are deleted specifically in primary afferent Nav1.8-positive neurons. Mutant animals were compared to controls for acute nociception, inflammatory pain, opiate-induced analgesia and constipation. There was a 76% decrease of mu receptor-positive neurons and a 60% reduction of mu-receptor mRNA in dorsal root ganglia of cKO mice. Mutant mice showed normal responses to heat, mechanical, visceral and chemical stimuli, as well as unchanged morphine antinociception and tolerance to antinociception in models of acute pain. Inflammatory pain developed similarly in cKO and controls mice after Complete Freund's Adjuvant. In the inflammation model, however, opiate-induced (morphine, fentanyl and loperamide) analgesia was reduced in mutant mice as compared to controls, and abolished at low doses. Morphine-induced constipation remained intact in cKO mice. We therefore genetically demonstrate for the first time that mu opioid receptors partly mediate opiate analgesia at the level of Nav1.8-positive sensory neurons. In our study, this mechanism operates under conditions of inflammatory pain, but not nociception. Previous pharmacology suggests that peripheral opiates may be clinically useful, and our data further demonstrate that Nav1.8 neuron-associated mu opioid receptors are feasible targets to alleviate some forms of persistent pain.


Subject(s)
Analgesia , Analgesics, Opioid/pharmacology , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Receptors, Opioid, mu/genetics , Animals , Constipation/chemically induced , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Female , Gene Deletion , Gene Expression , Gene Knockout Techniques , Gene Order , Gene Targeting , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Male , Mice , Mice, Knockout , Morphine/adverse effects , Morphine/pharmacology , NAV1.8 Voltage-Gated Sodium Channel/genetics , Nociception/drug effects , Pain/drug therapy , Pain/genetics , Pain Measurement , Protein Binding , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Sensory Thresholds/drug effects
17.
Neuropharmacology ; 75: 164-71, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23911743

ABSTRACT

Mammalian RF-amide peptides are encoded by five different genes and act through five different G protein-coupled receptors. RF-amide-related peptides-1 and -3, neuropeptides AF and FF, Prolactin releasing peptides, Kisspeptins and RFa peptides are currently considered endogenous peptides for NPFF1, NPFF2, GPR10, GPR54 and GPR103 receptors, respectively. However, several studies suggest that the selectivity of these peptides for their receptors is low and indicate that expression patterns for receptors and their corresponding ligands only partially overlap. In this study, we took advantage of the cloning of the five human RF-amide receptors to systematically examine their affinity for and their activation by all human RF-amide peptides. Binding experiments, performed on membranes from CHO cells expressing GPR10, GPR54 and GPR103 receptors, confirmed their high affinity and remarkable selectivity for their cognate ligands. Conversely, NPFF1 and NPFF2 receptors displayed high affinity for all RF-amide peptides. Moreover, GTPγS and cAMP experiments showed that almost all RF-amide peptides efficiently activate NPFF1 and NPFF2 receptors. As NPFF is known to modulate morphine analgesia, we undertook a systematic analysis in mice of the hyperalgesic and anti morphine-induced analgesic effects of a representative set of endogenous RF-amide peptides. All of them induced hyperalgesia and/or prevented morphine analgesia following intracerebroventricular administration. Importantly, these effects were prevented by administration of RF9, a highly selective NPFF1/NPFF2 antagonist. Altogether, our results show that all endogenous RF-amide peptides display pain-modulating properties and point to NPFF receptors as essential players for these effects.


Subject(s)
Analgesics, Opioid/pharmacology , Kisspeptins/metabolism , Morphine/pharmacology , Neuropeptides/metabolism , Prolactin-Releasing Hormone/metabolism , Receptors, Neuropeptide/metabolism , Animals , CHO Cells , Calcium/metabolism , Cricetulus , Cyclic AMP/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Humans , Kisspeptins/genetics , Male , Mice , Mice, Inbred C57BL , Neuropeptides/genetics , Pain Threshold/drug effects , Prolactin-Releasing Hormone/genetics , Protein Binding/drug effects , Time Factors , Tritium/pharmacokinetics
18.
Neuropsychopharmacology ; 38(6): 1050-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23303070

ABSTRACT

Pharmacological data suggest that delta opioid receptors modulate learning and memory processes. In the present study, we investigated whether inactivation of the delta opioid receptor modifies hippocampus (HPC)- and striatum-dependent behaviors. We first assessed HPC-dependent learning in mice lacking the receptor (Oprd1(-/-) mice) or wild-type (WT) mice treated with the delta opioid antagonist naltrindole using novel object recognition, and a dual-solution cross-maze task. Second, we subjected mutant animals to memory tests addressing striatum-dependent learning using a single-solution response cross-maze task and a motor skill-learning task. Genetic and pharmacological inactivation of delta opioid receptors reduced performance in HPC-dependent object place recognition. Place learning was also altered in Oprd1(-/-) animals, whereas striatum-dependent response and procedural learning were facilitated. Third, we investigated the expression levels for a large set of genes involved in neurotransmission in both HPC and striatum of Oprd1(-/-) mice. Gene expression was modified for several key genes that may contribute to alter hippocampal and striatal functions, and bias striatal output towards striatonigral activity. To test this hypothesis, we finally examined locomotor effects of dopamine receptor agonists. We found that Oprd1(-/-) and naltrindole-treated WT mice were more sensitive to the stimulant locomotor effect of SKF-81297 (D1/D5), supporting the hypothesis of facilitated striatonigral output. These data suggest, for the first time, that delta receptor activity tonically inhibits striatal function, and demonstrate that delta opioid receptors modulate learning and memory performance by regulating the HPC/striatum balance.


Subject(s)
Corpus Striatum/physiology , Hippocampus/physiology , Memory/physiology , Motor Activity/physiology , Motor Skills/physiology , Receptors, Opioid, delta/deficiency , Animals , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Receptors, Opioid, delta/genetics
19.
Addict Biol ; 17(1): 1-12, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21955143

ABSTRACT

Addiction is a chronic brain disorder. Prolonged abstinence from drugs of abuse involves dysphoria, high stress responsiveness and craving. The neurobiology of drug abstinence, however, is poorly understood. We previously identified a unique set of hundred mu-opioid receptor-dependent genes in the extended amygdala, a key site for hedonic and stress processing in the brain. Here we examined these candidate genes either immediately after chronic morphine, nicotine, Δ9-tetrahydrocannabinol or alcohol, or following 4 weeks of abstinence. Regulation patterns strongly differed among chronic groups. In contrast, gene regulations strikingly converged in the abstinent groups and revealed unforeseen common adaptations within a novel huntingtin-centered molecular network previously unreported in addiction research. This study demonstrates that, regardless the drug, a specific set of transcriptional regulations develops in the abstinent brain, which possibly contributes to the negative affect characterizing protracted abstinence. This transcriptional signature may represent a hallmark of drug abstinence and a unitary adaptive molecular mechanism in substance abuse disorders.


Subject(s)
Behavior, Addictive/physiopathology , Brain/drug effects , Gene Expression/drug effects , Gene Regulatory Networks/drug effects , Substance Withdrawal Syndrome/physiopathology , Substance-Related Disorders/physiopathology , Amygdala/drug effects , Animals , Behavior, Addictive/genetics , Cluster Analysis , Disease Models, Animal , Dronabinol/administration & dosage , Ethanol/administration & dosage , Gene Expression/genetics , Gene Regulatory Networks/genetics , Male , Mice , Mice, Inbred C57BL , Morphine/administration & dosage , Nicotine/administration & dosage , Principal Component Analysis , Real-Time Polymerase Chain Reaction , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/genetics , Substance Withdrawal Syndrome/genetics , Substance-Related Disorders/genetics , Temperance , Time , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
20.
Addict Biol ; 16(4): 615-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21392173

ABSTRACT

Alcoholism is characterized by a progressive loss of control over ethanol intake. The purpose of this study was to identify transcriptional changes selectively associated with excessive ethanol drinking in dependent mice, as opposed to non-dependent mice maintaining a stable voluntary consumption or mice solely undergoing forced intoxication. We measured expression levels of 106 candidate genes in the extended amygdala, a key brain structure for the development of drug addiction. Cluster analysis identified 17 and 15 genes selectively induced or repressed, respectively, under conditions of excessive drinking. These genes belong to signaling pathways involved in neurotransmission and transcriptional regulation.


Subject(s)
Alcoholic Intoxication/genetics , Alcoholic Intoxication/physiopathology , Alcoholism/genetics , Alcoholism/physiopathology , Amygdala/physiopathology , Gene Expression Regulation/physiology , Transcription, Genetic/genetics , Animals , Genetic Association Studies , Male , Mice , Mice, Inbred C57BL , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/physiology , Signal Transduction/genetics , Synaptic Transmission/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...