Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Mol Neurobiol ; 59(3): 1896-1911, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35032317

ABSTRACT

Cocaine addiction is a complex pathology inducing long-term neuroplastic changes that, in turn, contribute to maladaptive behaviors. This behavioral dysregulation is associated with transcriptional reprogramming in brain reward circuitry, although the mechanisms underlying this modulation remain poorly understood. The endogenous cannabinoid system may play a role in this process in that cannabinoid mechanisms modulate drug reward and contribute to cocaine-induced neural adaptations. In this study, we investigated whether cocaine self-administration induces long-term adaptations, including transcriptional modifications and associated epigenetic processes. We first examined endocannabinoid gene expression in reward-related brain regions of the rat following self-administered (0.33 mg/kg intravenous, FR1, 10 days) cocaine injections. Interestingly, we found increased Cnr1 expression in several structures, including prefrontal cortex, nucleus accumbens, dorsal striatum, hippocampus, habenula, amygdala, lateral hypothalamus, ventral tegmental area, and rostromedial tegmental nucleus, with most pronounced effects in the hippocampus. Endocannabinoid levels, measured by mass spectrometry, were also altered in this structure. Chromatin immunoprecipitation followed by qPCR in the hippocampus revealed that two activating histone marks, H3K4Me3 and H3K27Ac, were enriched at specific endocannabinoid genes following cocaine intake. Targeting CB1 receptors using chromosome conformation capture, we highlighted spatial chromatin re-organization in the hippocampus, as well as in the nucleus accumbens, suggesting that destabilization of the chromatin may contribute to neuronal responses to cocaine. Overall, our results highlight a key role for the hippocampus in cocaine-induced plasticity and broaden the understanding of neuronal alterations associated with endocannabinoid signaling. The latter suggests that epigenetic modifications contribute to maladaptive behaviors associated with chronic drug use.


Subject(s)
Cannabinoids , Cocaine , Animals , Cannabinoids/pharmacology , Cocaine/pharmacology , Hippocampus/metabolism , Male , Nucleus Accumbens/metabolism , Rats , Receptors, Cannabinoid/metabolism , Self Administration
2.
Eur J Nutr ; 60(8): 4621-4633, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34165614

ABSTRACT

OBJECTIVES: Increased availability of high-calorie palatable food in most countries has resulted in overconsumption of these foods, suggesting that excessive eating is driven by pleasure, rather than metabolic need. The behavior contributes to the rise in eating disorders, obesity, and associated pathologies like diabetes, cardiac disease, and cancers. The mesocorticolimbic dopamine and homeostatic circuits are interconnected and play a central role in palatable food intake. The endocannabinoid system is expressed in these circuits and represents a potent regulator of feeding, but the impact of an obesogenic diet on its expression is not fully known. METHODS: Food intake and body weight were recorded in male Wistar rats over a 6-week free-choice regimen of high fat and sugar; transcriptional regulations of the endocannabinoid system were examined post-mortem in brain reward regions (prefrontal cortex, nucleus accumbens, ventral tegmental area, and arcuate nucleus). K-means cluster analysis was used to classify animals based on individual sensitivity to obesity and palatable food intake. Endocannabinoid levels were quantified in the prefrontal cortex and nucleus accumbens. Gene expression in dopamine and homeostatic systems, including ghrelin and leptin receptors, and classical homeostatic peptides, were also investigated. RESULTS: The free-choice high-fat -and sugar diet induced hyperphagia and obesity in rats. Cluster analysis revealed that the propensity to develop obesity and excessive palatable food intake was differently associated with dopamine and endocannabinoid system gene expression in reward and homeostatic brain regions. CB2 receptor mRNA was increased in the nucleus accumbens of high sugar consumers, whereas CB1 receptor mRNA was decreased in obesity prone rats. CONCLUSIONS: Transcriptional data are consistent with observations of altered dopamine function in rodents that have access to an obesogenic diet and point to cannabinoid receptors as GPCR targets involved in neuroplasticity mechanisms associated with maladaptive intake of palatable food.


Subject(s)
Diet , Endocannabinoids , Animals , Brain , Cluster Analysis , Eating , Male , Obesity/etiology , Rats , Rats, Wistar , Reward
3.
Appetite ; 164: 105258, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33864862

ABSTRACT

Binge eating, the defining feature of binge eating disorder (BED), is associated with a number of adverse health outcomes as well as a reduced quality of life. Animals, like humans, selectively binge on highly palatable food suggesting that the behaviour is driven by hedonic, rather than metabolic, signals. Given the links to both reward processing and food intake, this study examined the contribution of the endocannabinoid system (ECS) to binge-like eating in rats. Separate groups were given intermittent (12 h) or continuous (24 h) access to 10% sucrose and food over 28 days, with only the 12 h access group displaying excessive sucrose intake within a discrete period of time (i.e., binge eating). Importantly, this group also exhibited alterations in ECS transcripts and endocannabinoid levels in brain reward regions, including an increase in cannabinoid receptor 1 (CB1R) mRNA in the nucleus accumbens as well as changes in endocannabinoid levels in the prefrontal cortex and hippocampus. We then tested whether different doses (1 and 3 mg/kg) of a CB1R antagonist, Rimonabant, modify binge-like intake or the development of a conditioned place preference (CPP) to sucrose. CB1R blockade reduced binge-like intake of sucrose and blocked a sucrose CPP, but only in rats that had undergone 28 days of sucrose consumption. These findings indicate that sucrose bingeing alters the ECS in reward-related areas, modifications that exacerbate the effect of CB1R blockade on sucrose reward. Overall, our results broaden the understanding of neural alterations associated with bingeing eating and demonstrate an important role for CB1R mechanisms in reward processing. In addition, these findings have implications for understanding substance abuse, which is also characterized by excessive and maladaptive intake, pointing towards addictive-like properties of palatable food.


Subject(s)
Binge-Eating Disorder , Animals , Eating , Endocannabinoids , Feeding Behavior , Quality of Life , Rats , Sucrose
4.
Eur J Neurosci ; 53(10): 3341-3349, 2021 05.
Article in English | MEDLINE | ID: mdl-33811699

ABSTRACT

Cocaine addiction is a complex pathology induced by long-term brain changes. Understanding the neurochemical changes underlying the reinforcing effects of this drug of abuse is critical for reducing the societal burden of drug addiction. The mu opioid receptor plays a major role in drug reward. This receptor is modulated by chronic cocaine treatment in specific brain structures, but few studies investigated neurochemical adaptations induced by voluntary cocaine intake. In this study, we investigated whether intravenous cocaine-self administration (0.33 mg/kg/injection, fixed-ratio 1 [FR1], 10 days) in rats induces transcriptional and functional changes of the mu opioid receptor in reward-related brain regions. Epigenetic processes with histone modifications were examined for two activating marks, H3K4Me3, and H3K27Ac. We found an increase of mu opioid receptor gene expression along with a potentiation of its functionality in hippocampus of cocaine self-administering animals compared to saline controls. Chromatin immunoprecipitation followed by qPCR revealed no modifications of the histone mark H3K4Me3 and H3K27Ac levels at mu opioid receptor promoter. Our study highlights the hippocampus as an important target to further investigate neuroadaptive processes leading to cocaine addiction.


Subject(s)
Cocaine , Animals , Hippocampus/metabolism , Rats , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Reward , Self Administration
5.
Neuropsychopharmacology ; 46(5): 920-927, 2021 04.
Article in English | MEDLINE | ID: mdl-33531622

ABSTRACT

The mu opioid receptor (MOR) is a G protein-coupled receptor that plays an essential role in reward and hedonic processes, and that has been implicated in disorders such as depression and addiction. Over the last decade, several brain imaging studies in depressed patients have consistently found that dysregulation of MOR function occurs in particular in the anterior insular cortex, an important brain site for the perception of internal states and emotional regulation. To investigate molecular mechanisms that may underlie these effects, here we assessed genetic polymorphisms, expression, and functional G-protein coupling of MOR in a large post-mortem cohort (N = 95) composed of depressed individuals who died by suicide, and healthy controls. Results indicated that depression, but not comorbid substance use disorder or acute opiate consumption, was associated with increased MOR activity. This effect was partly explained by a specific increase in expression of the inhibitory alpha G-protein subunit GNAI2. Consistent with previous neuroimaging studies, our findings support the notion that enhanced endogenous opioidergic tone in the anterior insula may buffer negative affective states in depressed individuals, a mechanism that could potentially contribute to the antidepressant efficacy of emerging opioid-based medications.


Subject(s)
Brain , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Receptors, Opioid, mu , Analgesics, Opioid , Brain/metabolism , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Emotions , Humans , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism
6.
Neurosci Lett ; 764: 135603, 2021 11 01.
Article in English | MEDLINE | ID: mdl-33387661

ABSTRACT

Cocaine addiction is a serious health issue in Western countries. Despite the regular increase in cocaine consumption across the population, there is no specific treatment for cocaine addiction. Critical roles for glutamate neurotransmission in the rewarding effects of psychostimulants as well as relapse have been suggested and accumulating evidence indicates that targeting mGlu group III receptors could represent a promising strategy to develop therapeutic compounds to treat addiction. In this context, the aim of our study was to examine the effect of LSP2-9166, a mGlu4/mGlu7 receptor orthosteric agonist, on the motivation for cocaine intake. We used an intravenous self-administration paradigm in male Wistar rats as a reliable model of voluntary drug intake. We first evaluated the direct impact of cocaine on Grm4 and Grm7 gene expression. Voluntary cocaine intake under a fixed ratio schedule of injections induced an increase of both mGlu4 and mGlu7 receptor transcripts in nucleus accumbens and hippocampus. We then evaluated the ability of LSP2-9166 to affect cocaine self-administration under a progressive ratio schedule of reinforcement. We found that this compound inhibits the motivation to obtain the drug, although it induced a hypolocomotor effect which could biais motivation index. Our findings demonstrate that mGlu group III receptors represent new targets for decreasing motivation to self-administer cocaine.


Subject(s)
Aminobutyrates/pharmacology , Cocaine-Related Disorders/drug therapy , Motivation/drug effects , Receptors, Metabotropic Glutamate/agonists , Administration, Intravenous , Aminobutyrates/therapeutic use , Animals , Cocaine/administration & dosage , Cocaine/adverse effects , Cocaine-Related Disorders/psychology , Disease Models, Animal , Glutamic Acid/metabolism , Humans , Male , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/metabolism , Reinforcement, Psychology , Self Administration , Synaptic Transmission/drug effects
7.
Biol Psychiatry ; 77(4): 404-15, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25444168

ABSTRACT

BACKGROUND: The delta opioid receptor (DOR) is broadly expressed throughout the nervous system; it regulates chronic pain, emotional responses, motivation, and memory. Neural circuits underlying DOR activities have been poorly explored by genetic approaches. We used conditional mouse mutagenesis to elucidate receptor function in GABAergic neurons of the forebrain. METHODS: We characterized DOR distribution in the brain of Dlx5/6-CreXOprd1(fl/fl) (Dlx-DOR) mice and tested main central DOR functions through behavioral testing. RESULTS: The DOR proteins were strongly deleted in olfactory bulb and striatum and remained intact in cortex and basolateral amygdala. Olfactory perception, circadian activity, and despair-like behaviors were unchanged. In contrast, locomotor stimulant effects of SNC80 (DOR agonist) and SKF81297 (D1 agonist) were abolished and increased, respectively. The Dlx-DOR mice showed lower levels of anxiety in the elevated plus maze, opposing the known high anxiety in constitutive DOR knockout animals. Also, Dlx-DOR mice reached the food more rapidly in a novelty suppressed feeding task, despite their lower motivation for food reward observed in an operant paradigm. Finally, c-fos protein staining after novelty suppressed feeding was strongly reduced in amygdala, concordant with the low anxiety phenotype of Dlx-DOR mice. CONCLUSIONS: We demonstrate that DORs expressed in the forebrain mediate the described locomotor effect of SNC80 and inhibit D1-stimulated hyperactivity. Our data also reveal an unanticipated anxiogenic role for this particular DOR subpopulation, with a potential novel adaptive role. In emotional responses, DORs exert dual anxiolytic and anxiogenic roles, both of which may have implications in the area of anxiety disorders.


Subject(s)
Anxiety/physiopathology , GABAergic Neurons/metabolism , Prosencephalon/metabolism , Receptors, Opioid, delta/metabolism , Animals , Behavior, Animal/physiology , Benzamides/pharmacology , Benzazepines/pharmacology , Brain/metabolism , Corpus Striatum/metabolism , Dopamine Agonists/pharmacology , Female , Male , Mice , Mice, Knockout , Motivation/physiology , Motor Activity/drug effects , Olfactory Bulb/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Receptors, Dopamine D1/agonists , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/analysis , Receptors, Opioid, delta/genetics
8.
Brain Struct Funct ; 220(2): 677-702, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24623156

ABSTRACT

Opioid receptors are G protein-coupled receptors (GPCRs) that modulate brain function at all levels of neural integration, including autonomic, sensory, emotional and cognitive processing. Mu (MOR) and delta (DOR) opioid receptors functionally interact in vivo, but whether interactions occur at circuitry, cellular or molecular levels remains unsolved. To challenge the hypothesis of MOR/DOR heteromerization in the brain, we generated redMOR/greenDOR double knock-in mice and report dual receptor mapping throughout the nervous system. Data are organized as an interactive database offering an opioid receptor atlas with concomitant MOR/DOR visualization at subcellular resolution, accessible online. We also provide co-immunoprecipitation-based evidence for receptor heteromerization in these mice. In the forebrain, MOR and DOR are mainly detected in separate neurons, suggesting system-level interactions in high-order processing. In contrast, neuronal co-localization is detected in subcortical networks essential for survival involved in eating and sexual behaviors or perception and response to aversive stimuli. In addition, potential MOR/DOR intracellular interactions within the nociceptive pathway offer novel therapeutic perspectives.


Subject(s)
Brain/metabolism , Nerve Net/metabolism , Neurons/metabolism , Receptors, Opioid, delta/analysis , Receptors, Opioid, mu/analysis , Animals , Female , Gene Knock-In Techniques , Male , Mice , Mice, Inbred C57BL
9.
Neuropsychopharmacology ; 39(11): 2694-705, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24874714

ABSTRACT

Addiction is a chronic disorder involving recurring intoxication, withdrawal, and craving episodes. Escaping this vicious cycle requires maintenance of abstinence for extended periods of time and is a true challenge for addicted individuals. The emergence of depressive symptoms, including social withdrawal, is considered a main cause for relapse, but underlying mechanisms are poorly understood. Here we establish a mouse model of protracted abstinence to heroin, a major abused opiate, where both emotional and working memory deficits unfold. We show that delta and kappa opioid receptor (DOR and KOR, respectively) knockout mice develop either stronger or reduced emotional disruption during heroin abstinence, establishing DOR and KOR activities as protective and vulnerability factors, respectively, that regulate the severity of abstinence. Further, we found that chronic treatment with the antidepressant drug fluoxetine prevents emergence of low sociability, with no impact on the working memory deficit, implicating serotonergic mechanisms predominantly in emotional aspects of abstinence symptoms. Finally, targeting the main serotonergic brain structure, we show that gene knockout of mu opioid receptors (MORs) in the dorsal raphe nucleus (DRN) before heroin exposure abolishes the development of social withdrawal. This is the first result demonstrating that intermittent chronic MOR activation at the level of DRN represents an essential mechanism contributing to low sociability during protracted heroin abstinence. Altogether, our findings reveal crucial and distinct roles for all three opioid receptors in the development of emotional alterations that follow a history of heroin exposure and open the way towards understanding opioid system-mediated serotonin homeostasis in heroin abuse.


Subject(s)
Heroin Dependence/physiopathology , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Social Behavior , Substance Withdrawal Syndrome/physiopathology , Animals , Antidepressive Agents, Second-Generation/pharmacology , Depression/metabolism , Disease Models, Animal , Dorsal Raphe Nucleus/drug effects , Dorsal Raphe Nucleus/metabolism , Fluoxetine/pharmacology , Heroin/pharmacology , Heroin Dependence/psychology , Male , Memory Disorders/physiopathology , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Narcotics/pharmacology , Receptors, Opioid, kappa/genetics , Receptors, Opioid, mu/genetics , Spatial Memory/drug effects , Spatial Memory/physiology , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/psychology
10.
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
11.
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
12.
Nat Neurosci ; 14(8): 1017-22, 2011 Jul 03.
Article in English | MEDLINE | ID: mdl-21725314

ABSTRACT

Zinc is abundant in the central nervous system and regulates pain, but the underlying mechanisms are unknown. In vitro studies have shown that extracellular zinc modulates a plethora of signaling membrane proteins, including NMDA receptors containing the NR2A subunit, which display exquisite zinc sensitivity. We created NR2A-H128S knock-in mice to investigate whether Zn2+-NR2A interaction influences pain control. In these mice, high-affinity (nanomolar) zinc inhibition of NMDA currents was lost in the hippocampus and spinal cord. Knock-in mice showed hypersensitivity to radiant heat and capsaicin, and developed enhanced allodynia in inflammatory and neuropathic pain models. Furthermore, zinc-induced analgesia was completely abolished under both acute and chronic pain conditions. Our data establish that zinc is an endogenous modulator of excitatory neurotransmission in vivo and identify a new mechanism in pain processing that relies on NR2A NMDA receptors. The study also potentially provides a molecular basis for the pain-relieving effects of dietary zinc supplementation.


Subject(s)
Neurons/drug effects , Pain/drug therapy , Receptors, N-Methyl-D-Aspartate/metabolism , Trace Elements/pharmacology , Acoustic Stimulation , Analysis of Variance , Animals , DNA Mutational Analysis , Disease Models, Animal , Dose-Response Relationship, Drug , Hand Strength/physiology , Hippocampus/cytology , Histidine/genetics , In Vitro Techniques , Larva , Locomotion/drug effects , Locomotion/genetics , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Pain/etiology , Pain/physiopathology , Pain Measurement , Pain Threshold/drug effects , Pain Threshold/physiology , Physical Stimulation , Protein Binding/drug effects , Reaction Time/drug effects , Reaction Time/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Reflex/drug effects , Rotarod Performance Test/methods , Serine/genetics , Smell/drug effects , Smell/genetics , Spinal Cord/cytology , Statistics, Nonparametric , Touch Perception/drug effects , Touch Perception/genetics , Trace Elements/therapeutic use , Xenopus , Zinc/pharmacology , Zinc/therapeutic use
13.
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
14.
Pain ; 152(6): 1238-1248, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21295407

ABSTRACT

Opioid receptors are major actors in pain control and are broadly distributed throughout the nervous system. A major challenge in pain research is the identification of key opioid receptor populations within nociceptive pathways, which control physiological and pathological pain. In particular, the respective contribution of peripheral vs. central receptors remains unclear, and it has not been addressed by genetic approaches. To investigate the contribution of peripheral delta opioid receptors in pain control, we created conditional knockout mice where delta receptors are deleted specifically in peripheral Na(V)1.8-positive primary nociceptive neurons. Mutant mice showed normal pain responses to acute heat and to mechanical and formalin stimuli. In contrast, mutant animals showed a remarkable increase of mechanical allodynia under both inflammatory pain induced by complete Freund adjuvant and neuropathic pain induced by partial sciatic nerve ligation. In these 2 models, heat hyperalgesia was virtually unchanged. SNC80, a delta agonist administered either systemically (complete Freund adjuvant and sciatic nerve ligation) or into a paw (sciatic nerve ligation), reduced thermal hyperalgesia and mechanical allodynia in control mice. However, these analgesic effects were absent in conditional mutant mice. In conclusion, this study reveals the existence of delta opioid receptor-mediated mechanisms, which operate at the level of Na(V)1.8-positive nociceptive neurons. Delta receptors in these neurons tonically inhibit mechanical hypersensitivity in both inflammatory and neuropathic pain, and they are essential to mediate delta opioid analgesia under conditions of persistent pain. This delta receptor population represents a feasible therapeutic target to alleviate chronic pain while avoiding adverse central effects. The conditional knockout of delta-opioid receptor in primary afferent Na(V)1.8 neurons augmented mechanical allodynia in persistent pain models and abolished delta opioid analgesia in these models.


Subject(s)
Ganglia, Spinal/pathology , Nociceptors/physiology , Pain/genetics , Pain/pathology , Receptors, Opioid, delta/deficiency , Analgesics, Opioid/therapeutic use , Analysis of Variance , Animals , Benzamides/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Freund's Adjuvant/adverse effects , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics , Inflammation/chemically induced , Inflammation/complications , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/genetics , NAV1.8 Voltage-Gated Sodium Channel , Nociceptors/drug effects , Pain/etiology , Pain Measurement/methods , Piperazines/therapeutic use , Protein Binding/drug effects , Protein Binding/genetics , Sodium Channels/genetics , Sodium Channels/metabolism , Sulfur Isotopes/pharmacokinetics
15.
J Neurosci ; 30(49): 16459-68, 2010 Dec 08.
Article in English | MEDLINE | ID: mdl-21147985

ABSTRACT

δ-Opioid receptors are G-protein-coupled receptors that regulate nociceptive and emotional responses. It has been well established that distinct agonists acting at the same G-protein-coupled receptor can engage different signaling or regulatory responses. This concept, known as biased agonism, has important biological and therapeutic implications. Ligand-biased responses are well described in cellular models, however, demonstrating the physiological relevance of biased agonism in vivo remains a major challenge. The aim of this study was to investigate the long-term consequences of ligand-biased trafficking of the δ-opioid receptor, at both the cellular and behavioral level. We used δ agonists with similar binding and analgesic properties, but high [SNC80 ((+)-4-[(αR)-α-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide)]- or low [ARM390 (N,N-diethyl-4-(phenyl-piperidin-4-ylidenemethyl)-benzamide)]-internalization potencies. As we found previously, a single SNC80-but not ARM390-administration triggered acute desensitization of the analgesic response in mice. However, daily injections of either compound over 5 d produced full analgesic tolerance. SNC80-tolerant animals showed widespread receptor downregulation, and tolerance to analgesic, locomotor and anxiolytic effects of the agonist. Hence, internalization-dependent tolerance developed, as a result of generalized receptor degradation. In contrast, ARM390-tolerant mice showed intact receptor expression, but δ-opioid receptor coupling to Ca²+ channels was abolished in dorsal root ganglia. Concomitantly, tolerance developed for agonist-induced analgesia, but not locomotor or anxiolytic responses. Therefore, internalization-independent tolerance was produced by anatomically restricted adaptations leading to pain-specific tolerance. Hence, ligand-directed receptor trafficking of the δ-opioid receptor engages distinct adaptive responses, and this study reveals a novel aspect of biased agonism in vivo.


Subject(s)
Analgesics/pharmacology , Drug Tolerance/physiology , Ligands , Pain Threshold/physiology , Receptors, Opioid, delta/metabolism , Analgesics/therapeutic use , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Brain/ultrastructure , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/genetics , Disease Models, Animal , Drug Interactions , Drug Tolerance/genetics , Female , Freund's Adjuvant , Ganglia, Spinal/cytology , Green Fluorescent Proteins/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Inflammation/chemically induced , Inflammation/complications , Locomotion/drug effects , Locomotion/genetics , Male , Maze Learning/drug effects , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pain/drug therapy , Pain/etiology , Pain Threshold/drug effects , Patch-Clamp Techniques/methods , Piperazines/pharmacology , Piperazines/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Protein Binding/drug effects , Protein Transport/genetics , Protein Transport/physiology , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/genetics , Sensory Receptor Cells/drug effects , Spinal Cord/ultrastructure , Statistics, Nonparametric , Sulfur Isotopes/metabolism , Time Factors
16.
PLoS One ; 4(5): e5425, 2009.
Article in English | MEDLINE | ID: mdl-19412545

ABSTRACT

BACKGROUND: GPCRs regulate a remarkable diversity of biological functions, and are thus often targeted for drug therapies. Stimulation of a GPCR by an extracellular ligand triggers receptor signaling via G proteins, and this process is highly regulated. Receptor activation is typically accompanied by desensitization of receptor signaling, a complex feedback regulatory process of which receptor internalization is postulated as a key event. The in vivo significance of GPCR internalization is poorly understood. In fact, the majority of studies have been performed in transfected cell systems, which do not adequately model physiological environments and the complexity of integrated responses observed in the whole animal. METHODS AND FINDINGS: In this study, we used knock-in mice expressing functional fluorescent delta opioid receptors (DOR-eGFP) in place of the native receptor to correlate receptor localization in neurons with behavioral responses. We analyzed the pain-relieving effects of two delta receptor agonists with similar signaling potencies and efficacies, but distinct internalizing properties. An initial treatment with the high (SNC80) or low (AR-M100390) internalizing agonist equally reduced CFA-induced inflammatory pain. However, subsequent drug treatment produced highly distinct responses. Animals initially treated with SNC80 showed no analgesic response to a second dose of either delta receptor agonist. Concomitant receptor internalization and G-protein uncoupling were observed throughout the nervous system. This loss of function was temporary, since full DOR-eGFP receptor responses were restored 24 hours after SNC80 administration. In contrast, treatment with AR-M100390 resulted in retained analgesic response to a subsequent agonist injection, and ex vivo analysis showed that DOR-eGFP receptor remained G protein-coupled on the cell surface. Finally SNC80 but not AR-M100390 produced DOR-eGFP phosphorylation, suggesting that the two agonists produce distinct active receptor conformations in vivo which likely lead to differential receptor trafficking. CONCLUSIONS: Together our data show that delta agonists retain full analgesic efficacy when receptors remain on the cell surface. In contrast, delta agonist-induced analgesia is abolished following receptor internalization, and complete behavioral desensitization is observed. Overall these results establish that, in the context of pain control, receptor localization fully controls receptor function in vivo. This finding has both fundamental and therapeutic implications for slow-recycling GPCRs.


Subject(s)
Behavior, Animal/drug effects , Behavior, Animal/physiology , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/physiology , Animals , Benzamides/pharmacology , Biological Transport, Active/drug effects , Cell Membrane/metabolism , Cells, Cultured , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Ligands , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/physiology , Pain/drug therapy , Pain/physiopathology , Phosphorylation , Piperazines/pharmacology , Piperidines/pharmacology , Protein Conformation , Receptors, Opioid, delta/chemistry , Receptors, Opioid, delta/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
17.
Neuropharmacology ; 54(3): 475-86, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18082850

ABSTRACT

Acute morphine administration produces analgesia and reward, but prolonged use may lead to analgesic tolerance in patients chronically treated for pain and to compulsive intake in opioid addicts. Moreover, long-term exposure may induce physical dependence, manifested as somatic withdrawal symptoms in the absence of the drug. We set up three behavioral paradigms to model these adaptations in mice, using distinct regimens of repeated morphine injections to induce either analgesic tolerance, locomotor sensitization or physical dependence. Interestingly, mice tolerant to analgesia were not sensitized to hyperlocomotion, whereas sensitized mice displayed some analgesic tolerance. We then examined candidate molecular modifications that could underlie the development of each behavioral adaptation. First, analgesic tolerance was not accompanied by mu opioid receptor desensitization in the periaqueductal gray. Second, cdk5 and p35 protein levels were unchanged in caudate-putamen, nucleus accumbens and prefrontal cortex of mice displaying locomotor sensitization. Finally, naloxone-precipitated morphine withdrawal did not enhance basal or forskolin-stimulated adenylate cyclase activity in nucleus accumbens, prefrontal cortex, amygdala, bed nucleus of stria terminalis or periaqueductal gray. Therefore, the expression of behavioral adaptations to chronic morphine treatment was not associated with the regulation of micro opioid receptor, cdk5 or adenylate cyclase activity in relevant brain areas. Although we cannot exclude that these modifications were not detected under our experimental conditions, another hypothesis is that alternative molecular mechanisms, yet to be discovered, underlie analgesic tolerance, locomotor sensitization and physical dependence induced by chronic morphine administration.


Subject(s)
Adenylyl Cyclases/metabolism , Cyclin-Dependent Kinase 5/metabolism , Drug Tolerance/physiology , Locomotion/drug effects , Morphine Dependence/etiology , Morphine/administration & dosage , Narcotics/administration & dosage , Receptors, Opioid, mu/metabolism , Analgesics , Animals , Behavior, Animal/drug effects , Brain/anatomy & histology , Brain/drug effects , Brain/metabolism , Drug Administration Schedule , Female , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Male , Mice , Mice, Inbred C57BL , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Protein Binding/drug effects , Time Factors
18.
Proc Natl Acad Sci U S A ; 103(25): 9691-6, 2006 Jun 20.
Article in English | MEDLINE | ID: mdl-16766653

ABSTRACT

The combination of fluorescent genetically encoded proteins with mouse engineering provides a fascinating means to study dynamic biological processes in mammals. At present, green fluorescent protein (GFP) mice were mainly developed to study gene expression patterns or cell morphology and migration. Here we used enhanced GFP (EGFP) to achieve functional imaging of a G protein-coupled receptor (GPCR) in vivo. We created mice where the delta-opioid receptor (DOR) is replaced by an active DOR-EGFP fusion. Confocal imaging revealed detailed receptor neuroanatomy throughout the nervous system of knock-in mice. Real-time imaging in primary neurons allowed dynamic visualization of drug-induced receptor trafficking. In DOR-EGFP animals, drug treatment triggered receptor endocytosis that correlated with the behavioral response. Mice with internalized receptors were insensitive to subsequent agonist administration, providing evidence that receptor sequestration limits drug efficacy in vivo. Direct receptor visualization in mice is a unique approach to receptor biology and drug design.


Subject(s)
Green Fluorescent Proteins/metabolism , Receptors, Opioid, delta/genetics , Receptors, Opioid, delta/metabolism , Animals , Brain/metabolism , Endocytosis , Gene Expression , Green Fluorescent Proteins/genetics , Kinetics , Mice , Mice, Transgenic , Transgenes/genetics
19.
J Neurochem ; 97(1): 292-301, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16515546

ABSTRACT

The diversity of peptide ligands for a particular receptor may provide a greater dynamic range of functional responses, while maintaining selectivity in receptor activation. Dynorphin A (Dyn A), and dynorphin B (Dyn B) are endogenous opioid peptides that activate the kappa-opioid receptor (KOR). Here, we characterized interactions of big dynorphin (Big Dyn), a 32-amino acid prodynorphin-derived peptide consisting of Dyn A and Dyn B, with human KOR, mu- (hMOR) and delta- (hDOR) opioid receptors and opioid receptor-like receptor 1 (hORL1) expressed in cells transfected with respective cDNA. Big Dyn and Dyn A demonstrated roughly similar affinity for binding to hKOR that was higher than that of Dyn B. Dyn A was more selective for hKOR over hMOR, hDOR and hORL1 than Big Dyn, while Dyn B demonstrated low selectivity. In contrast, Big Dyn activated G proteins through KOR with much greater potency, efficacy and selectivity than other dynorphins. There was no correlation between the rank order of the potency for the KOR-mediated activation of G proteins and the binding affinity of dynorphins for KOR. The rank of the selectivity for the activation of G proteins through hKOR and of the binding to this receptor also differed. Immunoreactive Big Dyn was detected using the combination of radioimmunoassay (RIA) and HPLC in the human nucleus accumbens, caudate nucleus, hippocampus and cerebrospinal fluid (CSF) with the ratio of Big Dyn and Dyn B being approximately 1:3. The presence in the brain implies that Big Dyn, along with other dynorphins, is processed from prodynorphin and secreted from neurons. Collectively, the high potency and efficacy and the relative abundance suggest that Big Dyn may play a role in the KOR-mediated activation of G proteins.


Subject(s)
Binding, Competitive/physiology , Central Nervous System/metabolism , Dynorphins/cerebrospinal fluid , Receptors, Opioid, kappa/metabolism , Animals , Binding, Competitive/drug effects , Central Nervous System/drug effects , Cerebrospinal Fluid/metabolism , Dynorphins/chemistry , Dynorphins/genetics , Endorphins/cerebrospinal fluid , Endorphins/chemistry , Endorphins/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Ligands , Mice , Mice, Knockout , Neural Pathways/drug effects , Neural Pathways/metabolism , Neurons/drug effects , Neurons/metabolism , Pain/genetics , Pain/metabolism , Pain/physiopathology , Radioimmunoassay , Radioligand Assay , Receptors, Opioid/drug effects , Receptors, Opioid/metabolism , Receptors, Opioid, delta/drug effects , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism , Nociceptin Receptor
20.
Nat Struct Biol ; 10(8): 629-36, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12847517

ABSTRACT

The high resolution structure of rhodopsin has greatly enhanced current understanding of G protein-coupled receptor (GPCR) structure in the off-state, but the activation process remains to be clarified. We investigated molecular mechanisms of delta-opioid receptor activation without a preconceived structural hypothesis. Using random mutagenesis of the entire receptor, we identified 30 activating point mutations. Three-dimensional modeling revealed an activation path originating from the third extracellular loop and propagating through tightly packed helices III, VI and VII down to a VI-VII cytoplasmic switch. N- and C-terminal determinants also influence receptor activity. Findings for this therapeutically important receptor may apply to other GPCRs that respond to diffusible ligands.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Opioid, delta/chemistry , Receptors, Opioid, delta/genetics , Binding Sites , Cell Line , Humans , In Vitro Techniques , Models, Molecular , Mutagenesis , Point Mutation , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid, delta/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...