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










Publication year range
1.
Trends Neurosci ; 46(4): 257-259, 2023 04.
Article in English | MEDLINE | ID: mdl-36707259

ABSTRACT

The ventral tegmental area (VTA) has a pivotal role in motivated behavior. Much of the research on the VTA has focused on the mesocorticolimbic dopamine projections and their role in the computation of a 'reward prediction error' (RPE) for reward-guided learning. In a recent study, Zhou et al. report that VTA GABA neurons, the axons of which innervate the ventral pallidum (VP), have a unique role in signaling reward value to the basal ganglia and guiding reward seeking.


Subject(s)
Basal Ganglia , Ventral Tegmental Area , Humans , Ventral Tegmental Area/physiology , Dopamine , Reward
2.
Addict Biol ; 26(2): e12938, 2021 03.
Article in English | MEDLINE | ID: mdl-32666571

ABSTRACT

Our previous studies consistently showed that MDMA-induced locomotor hyperactivity is dramatically increased by coadministration of ethanol (EtOH) in rats, indicating possible potentiation of MDMA abuse liability. Thus, we aimed to identify the brain region(s) and neuropharmacological substrates involved in the pharmacodynamics of this potentiation. We first showed that potentiation of locomotor activity by the combination of ip administration of EtOH (1.5 g/kg) and MDMA (6.6 mg/kg) is delay sensitive and maximal when both drugs are injected simultaneously. Then, we used the 2-deoxyglucose quantitative autoradiography technique to assess the impact of EtOH, MDMA, or their combination on local cerebral metabolic rates for glucose (CMRglcs). We showed a specific metabolic activation in the ventral striatum (VS) under MDMA + EtOH versus MDMA or EtOH alone. We next tested if reversible (tetrodotoxin, TTX) or permanent (6-hydrodoxyopamine, 6-OHDA) lesion of the VS could affect locomotor response to MDMA and MDMA + EtOH. Finally, we blocked dopamine D1 or glutamate NMDA receptors in the VS and measured the effects of MDMA and MDMA + EtOH on locomotor activity. We showed that bilateral reversible inactivation (TTX) or permanent lesion (6-OHDA) of the VS prevented the potentiation by EtOH of MDMA-induced locomotor hyperactivity. Likewise, blockade of D1 or NMDA receptors in the VS also reduced the potentiation of MDMA locomotor activity by EtOH. These data indicate that dopamine D1 and glutamate NMDA receptor-driven mechanisms in the VS play a key role in the pharmacodynamics of EtOH-induced potentiation of the locomotor effects of MDMA.


Subject(s)
Ethanol/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Ventral Striatum/drug effects , Animals , Drug Combinations , Drug Synergism , Ethanol/administration & dosage , Locomotion/drug effects , Male , N-Methyl-3,4-methylenedioxyamphetamine/administration & dosage , Oxidopamine/pharmacology , Rats , Rats, Long-Evans , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Tetrodotoxin/pharmacology
3.
Article in English | MEDLINE | ID: mdl-32341062

ABSTRACT

Cocaine leads to a strong euphoria, which is at the origin of its recreational use. Past the acute effects, the drug leaves traces in the brain that persist long after it has been cleared from the body. These traces eventually shape behavior such that drug use may become compulsive, and addiction develops. Here, we discuss cocaine-evoked synaptic plasticity of glutamatergic transmission onto dopamine (DA) neurons of the ventral tegmental area (VTA) as one of the earliest traces after a first injection of cocaine. We review the literature that has examined the induction requirements, as well as the expression mechanism of this form of plasticity, and ask the question about its functional significance.


Subject(s)
Cocaine/pharmacokinetics , Neuronal Plasticity/drug effects , Synaptic Transmission/drug effects , Ventral Tegmental Area/drug effects , Humans , Substance-Related Disorders/etiology
4.
Neuron ; 107(1): 158-172.e4, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32333845

ABSTRACT

Overeating typically follows periods of energy deficit, but it is also sustained by highly palatable foods, even without metabolic demand. Dopamine D1 receptor-expressing medium spiny neurons (D1-MSNs) of the nucleus accumbens shell (NAcSh) project to the lateral hypothalamus (LH) to authorize feeding when inhibited. Whether plasticity at these synapses can affect food intake is unknown. Here, ex vivo electrophysiology recordings reveal that D1-MSN-to-LH inhibitory transmission is depressed in circumstances in which overeating is promoted. Endocannabinoid signaling is identified as the induction mechanism, since inhibitory plasticity and concomitant overeating were blocked or induced by CB1R antagonism or agonism, respectively. D1-MSN-to-LH projectors were largely non-overlapping with D1-MSNs targeting ventral pallidum or ventral midbrain, providing an anatomical basis for distinct circuit plasticity mechanisms. Our study reveals a critical role for plasticity at D1-MSN-to-LH synapses in adaptive feeding control, which may underlie persistent overeating of unhealthy foods, a major risk factor for developing obesity.


Subject(s)
Hyperphagia/physiopathology , Hypothalamic Area, Lateral/physiopathology , Long-Term Synaptic Depression/physiology , Nucleus Accumbens/physiopathology , Synaptic Transmission/physiology , Animals , Mice , Neural Pathways/physiopathology
5.
Science ; 364(6444): 991-995, 2019 Jun 07.
Article in English | MEDLINE | ID: mdl-31171697

ABSTRACT

When an animal is facing unfamiliar food, its odor, together with semiochemicals emanating from a conspecific, can constitute a safety message and authorize intake. The piriform cortex (PiC) codes olfactory information, and the inactivation of neurons in the nucleus accumbens (NAc) can acutely trigger consumption. However, the neural circuit and cellular substrate of transition of olfactory perception into value-based actions remain elusive. We detected enhanced activity after social transmission between two mice in neurons of the medial prefrontal cortex (mPFC) that target the NAc and receive projections from the PiC. Exposure to a conspecific potentiated the excitatory postsynaptic currents in NAc projectors, whereas blocking transmission from PiC to mPFC prevented social transmission. Thus, synaptic plasticity in the mPFC is a cellular substrate of social transmission of food safety.


Subject(s)
Food Preferences/psychology , Food Safety , Neuronal Plasticity/physiology , Piriform Cortex/physiology , Prefrontal Cortex/physiology , Social Behavior , Animals , Mice , Mice, Inbred C57BL
6.
Nature ; 564(7736): 366-371, 2018 12.
Article in English | MEDLINE | ID: mdl-30568192

ABSTRACT

Activation of the mesolimbic dopamine system reinforces goal-directed behaviours. With repetitive stimulation-for example, by chronic drug abuse-the reinforcement may become compulsive and intake continues even in the face of major negative consequences. Here we gave mice the opportunity to optogenetically self-stimulate dopaminergic neurons and observed that only a fraction of mice persevered if they had to endure an electric shock. Compulsive lever pressing was associated with an activity peak in the projection terminals from the orbitofrontal cortex (OFC) to the dorsal striatum. Although brief inhibition of OFC neurons temporarily relieved compulsive reinforcement, we found that transmission from the OFC to the striatum was permanently potentiated in persevering mice. To establish causality, we potentiated these synapses in vivo in mice that stopped optogenetic self-stimulation of dopamine neurons because of punishment; this led to compulsive lever pressing, whereas depotentiation in persevering mice had the converse effect. In summary, synaptic potentiation of transmission from the OFC to the dorsal striatum drives compulsive reinforcement, a defining symptom of addiction.


Subject(s)
Behavior, Addictive/physiopathology , Compulsive Behavior/physiopathology , Models, Neurological , Neuronal Plasticity , Animals , Behavior, Addictive/pathology , Behavior, Addictive/psychology , Compulsive Behavior/pathology , Compulsive Behavior/psychology , Dopaminergic Neurons/physiology , Electric Stimulation , Female , Male , Mice , Neostriatum/cytology , Neostriatum/physiology , Neural Inhibition , Neural Pathways , Optogenetics , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Punishment , Reinforcement, Psychology , Stochastic Processes , Synapses/metabolism , Synaptic Transmission
7.
Elife ; 72018 10 30.
Article in English | MEDLINE | ID: mdl-30373717

ABSTRACT

The dopamine (DA) hypothesis posits the increase of mesolimbic dopamine levels as a defining commonality of addictive drugs, initially causing reinforcement, eventually leading to compulsive consumption. While much experimental evidence from psychostimulants supports this hypothesis, it has been challenged for opioid reinforcement. Here, we monitor genetically encoded DA and calcium indicators as well as cFos in mice to reveal that heroin activates DA neurons located in the medial part of the VTA, preferentially projecting to the medial shell of the nucleus accumbens (NAc). Chemogenetic and optogenetic manipulations of VTA DA or GABA neurons establish a causal link to heroin reinforcement. Inhibition of DA neurons blocked heroin self-administration, while heroin inhibited optogenetic self-stimulation of DA neurons. Likewise, heroin occluded the self-inhibition of VTA GABA neurons. Together, these experiments support a model of disinhibition of a subset of VTA DA neurons in opioid reinforcement.


Subject(s)
Dopaminergic Neurons/physiology , Heroin/adverse effects , Nucleus Accumbens/physiology , Reinforcement, Psychology , Animals , Dopamine/metabolism , HEK293 Cells , Humans , Mice, Inbred C57BL , Optogenetics , Self Administration , Ventral Tegmental Area/physiology
9.
Cannabis Cannabinoid Res ; 2(1): 8-20, 2017.
Article in English | MEDLINE | ID: mdl-28861501

ABSTRACT

Introduction: The GPR55 receptor has been identified as an atypical cannabinoid receptor and is implicated in various physiological processes. However, its functional role in the central nervous system is not currently understood. The presence of GPR55 receptor in neural regions such as the ventral hippocampus (vHipp), which is critical for cognition, recognition memory, and affective processing, led us to hypothesize that intra-vHipp GPR55 transmission may modulate mesolimbic activity states and related behavioral phenomena. The vHipp is involved in contextual memory and affective regulation through functional interactions with the mesolimbic dopamine system. Materials and Methods: Using a combination of in vivo electrophysiology and behavioral pharmacological assays in rats, we tested whether intra-vHipp activation of GPR55 receptor transmission with the fatty acid amide, palmitoylethanolamide (PEA), a lipid neuromodulator with agonist actions at the GPR55 receptor, may modulate mesolimbic dopaminergic activity states. We further examined the potential effects of intra-vHipp PEA in affective, cognitive and contextual memory tasks. Discussion: We report that intra-vHipp PEA produces a hyper-dopaminergic state in the mesolimbic system characterized by increased firing and bursting activity of ventral tegmental area dopaminergic neuron populations. Furthermore, while PEA-induced activation of GPR55 transmission had no effects on opiate-related reward-related memory formation, we observed strong disruptions in social interaction and recognition memory, spatial location memory, and context-independent associative fear memory formation. Finally, the effects of intra-vHipp PEA were blocked by a selective GPR55 receptor antagonist, CID160 and were dependent upon NMDA receptor transmission, directly in the vHipp. Conclusions: The present results add to a growing body of evidence demonstrating important functional roles for GPR55 signaling in cannabinoid-related neuronal and behavioral phenomena and underscore the potential for GPR55 signaling in the mediation of cannabinoid-related effects independently of the CB1/CB2 receptor systems.

10.
Cereb Cortex ; 27(2): 1297-1310, 2017 02 01.
Article in English | MEDLINE | ID: mdl-26733534

ABSTRACT

Considerable evidence suggests that adolescent exposure to delta-9-tetrahydrocanabinol (THC), the psychoactive component in marijuana, increases the risk of developing schizophrenia-related symptoms in early adulthood. In the present study, we used a combination of behavioral and molecular analyses with in vivo neuronal electrophysiology to compare the long-term effects of adolescent versus adulthood THC exposure in rats. We report that adolescent, but not adult, THC exposure induces long-term neuropsychiatric-like phenotypes similar to those observed in clinical populations. Thus, adolescent THC exposure induced behavioral abnormalities resembling positive and negative schizophrenia-related endophenotypes and a state of neuronal hyperactivity in the mesocorticolimbic dopamine (DA) pathway. Furthermore, we observed profound alterations in several prefrontal cortical molecular pathways consistent with sub-cortical DAergic dysregulation. Our findings demonstrate a profound dissociation in relative risk profiles for adolescent versus adulthood exposure to THC in terms of neuronal, behavioral, and molecular markers resembling neuropsychiatric pathology.


Subject(s)
Cannabinoids/pharmacology , Dopamine/metabolism , Prefrontal Cortex/drug effects , Social Behavior , Aging , Animals , Behavior, Animal/drug effects , Cannabinoids/metabolism , Hyperkinesis/metabolism , Male , Prefrontal Cortex/metabolism , Rats, Sprague-Dawley , Schizophrenia/metabolism
11.
Article in English | MEDLINE | ID: mdl-27493155

ABSTRACT

BACKGROUND: Multiple studies suggest a pivotal role of the endocannabinoid system in regulating the reinforcing effects of various substances of abuse. Rimonabant, a CB1 inverse agonist found to be effective for smoking cessation, was associated with an increased risk of anxiety and depression. Here we evaluated the effects of the CB1 neutral antagonist AM4113 on the abuse-related effects of nicotine and its effects on anxiety and depressive-like behavior in rats. METHODS: Rats were trained to self-administer nicotine under a fixed-ratio 5 or progressive-ratio schedules of reinforcement. A control group was trained to self-administer food. The acute/chronic effects of AM4113 pretreatment were evaluated on nicotine taking, motivation for nicotine, and cue-, nicotine priming- and yohimbine-induced reinstatement of nicotine-seeking. The effects of AM4113 in the basal firing and bursting activity of midbrain dopamine neurons were evaluated in a separate group of animals treated with nicotine. Anxiety/depression-like effects of AM4113 and rimonabant were evaluated 24h after chronic (21 days) pretreatment (0, 1, 3, and 10mg/kg, 1/d). RESULTS: AM4113 significantly attenuated nicotine taking, motivation for nicotine, as well as cue-, priming- and stress-induced reinstatement of nicotine-seeking behavior. These effects were accompanied by a decrease of the firing and burst rates in the ventral tegmental area dopamine neurons in response to nicotine. On the other hand, AM4113 pretreatment did not have effects on operant responding for food. Importantly, AM4113 did not have effects on anxiety and showed antidepressant-like effects. CONCLUSION: Our results indicate that AM4113 could be a promising therapeutic option for the prevention of relapse to nicotine-seeking while lacking anxiety/depression-like side effects.


Subject(s)
Behavior, Addictive/drug therapy , Behavior, Animal/drug effects , Cannabinoid Receptor Antagonists/pharmacology , Mesencephalon/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Tobacco Use Cessation Devices , Tobacco Use Disorder/drug therapy , Weight Loss/drug effects , Animals , Anxiety/chemically induced , Anxiety/psychology , Behavior, Addictive/metabolism , Behavior, Addictive/physiopathology , Behavior, Addictive/psychology , Cannabinoid Receptor Antagonists/toxicity , Cues , Depression/chemically induced , Depression/psychology , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dose-Response Relationship, Drug , Drug Inverse Agonism , Drug-Seeking Behavior/drug effects , Male , Maze Learning/drug effects , Mesencephalon/metabolism , Mesencephalon/physiopathology , Motivation/drug effects , Motor Activity/drug effects , Piperidines/toxicity , Pyrazoles/toxicity , Rats, Long-Evans , Rats, Wistar , Receptor, Cannabinoid, CB1/metabolism , Rimonabant , Signal Transduction/drug effects , Swimming , Time Factors , Tobacco Use Disorder/metabolism , Tobacco Use Disorder/physiopathology , Tobacco Use Disorder/psychology
12.
Neuropsychopharmacology ; 41(12): 2839-2850, 2016 11.
Article in English | MEDLINE | ID: mdl-27296152

ABSTRACT

Emerging evidence suggests that the largest phytochemical component of cannabis, cannabidiol (CBD), may possess pharmacotherapeutic properties in the treatment of neuropsychiatric disorders. CBD has been reported to functionally interact with both the mesolimbic dopamine (DA) and serotonergic (5-HT) receptor systems. However, the underlying mechanisms by which CBD may modulate emotional processing are not currently understood. Using a combination of in vivo electrophysiological recording and fear conditioning in rats, the present study aimed to characterize the behavioral, neuroanatomical, and pharmacological effects of CBD within the mesolimbic pathway, and its possible functional interactions with 5-HT and DAergic transmission. Using targeted microinfusions of CBD into the shell region of the mesolimbic nucleus accumbens (NASh), we report that intra-NASh CBD potently blocks the formation of conditioned freezing behaviors. These effects were challenged with DAergic, cannabinoid CB1 receptor, and serotonergic (5-HT1A) transmission blockade, but only 5-HT1A blockade restored associative conditioned freezing behaviors. In vivo intra-ventral tegmental area (VTA) electrophysiological recordings revealed that behaviorally effective doses of intra-NASh CBD elicited a predominant decrease in spontaneous DAergic neuronal frequency and bursting activity. These neuronal effects were reversed by simultaneous blockade of 5-HT1A receptor transmission. Finally, using a functional contralateral disconnection procedure, we demonstrated that the ability of intra-NASh CBD to block the formation of conditioned freezing behaviors was dependent on intra-VTA GABAergic transmission substrates. Our findings demonstrate a novel NAcVTA circuit responsible for the behavioral and neuronal effects of CBD within the mesolimbic system via functional interactions with serotonergic 5-HT1A receptor signaling.


Subject(s)
Cannabidiol/pharmacology , Dopamine/metabolism , Fear/drug effects , Memory/drug effects , Neurons/drug effects , Nucleus Accumbens/cytology , Serotonin/metabolism , Synaptic Transmission/drug effects , Ventral Tegmental Area/cytology , Action Potentials/drug effects , Animals , Conditioning, Psychological/drug effects , Dopamine/pharmacology , Dose-Response Relationship, Drug , Drug Combinations , Electroshock , Male , Neural Pathways/physiology , Neurotransmitter Agents/pharmacology , Nucleus Accumbens/drug effects , Rats , Rats, Sprague-Dawley , Serotonin/pharmacology
13.
J Neurosci ; 36(18): 5160-9, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27147666

ABSTRACT

UNLABELLED: Schizophrenia-related psychosis is associated with disturbances in mesolimbic dopamine (DA) transmission, characterized by hyperdopaminergic activity in the mesolimbic pathway. Currently, the only clinically effective treatment for schizophrenia involves the use of antipsychotic medications that block DA receptor transmission. However, these medications produce serious side effects leading to poor compliance and treatment outcomes. Emerging evidence points to the involvement of a specific phytochemical component of marijuana called cannabidiol (CBD), which possesses promising therapeutic properties for the treatment of schizophrenia-related psychoses. However, the neuronal and molecular mechanisms through which CBD may exert these effects are entirely unknown. We used amphetamine (AMPH)-induced sensitization and sensorimotor gating in rats, two preclinical procedures relevant to schizophrenia-related psychopathology, combined with in vivo single-unit neuronal electrophysiology recordings in the ventral tegmental area, and molecular analyses to characterize the actions of CBD directly in the nucleus accumbens shell (NASh), a brain region that is the current target of most effective antipsychotics. We demonstrate that Intra-NASh CBD attenuates AMPH-induced sensitization, both in terms of DAergic neuronal activity measured in the ventral tegmental area and psychotomimetic behavioral analyses. We further report that CBD controls downstream phosphorylation of the mTOR/p70S6 kinase signaling pathways directly within the NASh. Our findings demonstrate a novel mechanism for the putative antipsychotic-like properties of CBD in the mesolimbic circuitry. We identify the molecular signaling pathways through which CBD may functionally reduce schizophrenia-like neuropsychopathology. SIGNIFICANCE STATEMENT: The cannabis-derived phytochemical, cannabidiol (CBD), has been shown to have pharmacotherapeutic efficacy for the treatment of schizophrenia. However, the mechanisms by which CBD may produce antipsychotic effects are entirely unknown. Using preclinical behavioral procedures combined with molecular analyses and in vivo neuronal electrophysiology, our findings identify a functional role for the nucleus accumbens as a critical brain region whereby CBD can produce effects similar to antipsychotic medications by triggering molecular signaling pathways associated with the effects of classic antipsychotic medications. Specifically, we report that CBD can attenuate both behavioral and dopaminergic neuronal correlates of mesolimbic dopaminergic sensitization, via a direct interaction with mTOR/p70S6 kinase signaling within the mesolimbic pathway.


Subject(s)
Amphetamine/antagonists & inhibitors , Behavior, Animal/drug effects , Cannabidiol/pharmacology , Central Nervous System Stimulants/antagonists & inhibitors , Dopamine Uptake Inhibitors/antagonists & inhibitors , Dopaminergic Neurons/drug effects , Limbic System/physiology , Neural Pathways/drug effects , Neurons/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/drug effects , TOR Serine-Threonine Kinases/drug effects , Amphetamine/pharmacology , Animals , Central Nervous System Stimulants/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Male , Motor Activity/drug effects , Nucleus Accumbens/drug effects , Rats , Rats, Sprague-Dawley , Reflex, Startle/drug effects
14.
Biol Psychiatry ; 80(3): 216-25, 2016 08 01.
Article in English | MEDLINE | ID: mdl-26681496

ABSTRACT

BACKGROUND: Cannabinoid receptor transmission strongly influences emotional processing, and disturbances in cannabinoid signaling are associated with various neuropsychiatric disorders. The mammalian ventral hippocampus (vHipp) is a critical neural region controlling mesolimbic activity via glutamatergic projections to the nucleus accumbens. Furthermore, vHipp abnormalities are linked to schizophrenia-related psychopathology. Nevertheless, the mechanisms by which intra-vHipp cannabinoid signaling may modulate mesolimbic activity states and emotional processing are not currently understood. METHODS: Using an integrative combination of in vivo electrophysiological recordings and behavioral pharmacologic assays in rats, we tested whether activation of cannabinoid type 1 receptors (CB1R) in the vHipp may modulate neuronal activity in the shell subregion of the nucleus accumbens (NASh). We next examined how vHipp CB1R signaling may control the salience of rewarding or aversive emotional memory formation and social interaction/recognition behaviors via intra-NASh glutamatergic transmission. RESULTS: We demonstrate for the first time that vHipp CB1R transmission can potently modulate NASh neuronal activity and can differentially control the formation of context-dependent and context-independent forms of rewarding or aversion-related emotional associative memories. In addition, we found that activation of vHipp CB1R transmission strongly disrupts normal social behavior and cognition. Finally, we report that these behavioral effects are dependent upon intra-NASh alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid/N-methyl-D-aspartate receptor transmission. CONCLUSIONS: Together, these findings demonstrate a critical role for hippocampal cannabinoid signaling in the modulation of mesolimbic neuronal activity states and suggest that dysregulation of CB1R transmission in the vHipp→NASh circuit may underlie hippocampal-mediated affective and social behavioral disturbances present in neuropsychiatric disorders.


Subject(s)
Avoidance Learning , Emotions/physiology , Hippocampus/physiology , Nucleus Accumbens/cytology , Nucleus Accumbens/physiology , Receptor, Cannabinoid, CB1/physiology , Reward , Animals , Benzoxazines/administration & dosage , Benzoxazines/pharmacology , Cognition/physiology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dizocilpine Maleate/administration & dosage , Dizocilpine Maleate/pharmacology , Hippocampus/drug effects , Male , Memory/physiology , Microinjections , Morpholines/administration & dosage , Morpholines/pharmacology , Naphthalenes/administration & dosage , Naphthalenes/pharmacology , Neurons/physiology , Nucleus Accumbens/drug effects , Piperidines/administration & dosage , Piperidines/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Quinoxalines/administration & dosage , Quinoxalines/pharmacology , Rats , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptors, AMPA/agonists , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/physiology , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Rimonabant , Social Behavior
15.
Brain Struct Funct ; 221(1): 91-102, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25260556

ABSTRACT

Systems-level consolidation models propose that recent memories are initially hippocampus-dependent. When remote, they are partially or completely dependent upon the medial prefrontal cortex (mPFC). An implication of the mPFC in recent memory, however, is still debated. Different amounts of muscimol (MSCI 0, 30, 50, 80 and 250 ng in 1 µL PBS) were used to assess the impact of inactivation of the dorsal hippocampus (dHip) or the mPFC (targeting the prelimbic cortex) on a 24-h delayed retrieval of a platform location that rats had learned drug-free in a water maze. The two smallest amounts of MSCI (30 and 50 ng) did not affect recall, whatever the region. 80 ng MSCI infused into the dHip disrupted spatial memory retrieval, as did the larger amount. Infusion of MSCI into the mPFC did not alter performance in the 0-80 ng range. At 250 ng, it induced an as dramatic memory impairment as after efficient dHip inactivation. Stereological quantifications showed that 80 ng MSCI in the dHip and 250 ng MSCI in the mPFC induced a more than 80% reduction of c-Fos expression, suggesting that, beyond the amounts infused, it is the magnitude of the neuronal activity decrease which is determinant as to the functional outcome of the inactivation. Because, based on the literature, even 250 ng MSCI is a small amount, our results point to a contribution of the mPFC to the recall of a recently acquired spatial memory and thereby extend our knowledge about the functions of this major actor of cognition.


Subject(s)
Hippocampus/physiology , Prefrontal Cortex/physiology , Spatial Memory/physiology , Animals , GABA-A Receptor Agonists/administration & dosage , Hippocampus/drug effects , Hippocampus/metabolism , Male , Maze Learning/drug effects , Maze Learning/physiology , Mental Recall/drug effects , Mental Recall/physiology , Muscimol/administration & dosage , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Long-Evans , Spatial Memory/drug effects
16.
Neuropsychopharmacology ; 40(6): 1436-47, 2015 May.
Article in English | MEDLINE | ID: mdl-25510937

ABSTRACT

Disturbances in cannabinoid type 1 receptor (CB1R) signaling have been linked to emotional and cognitive deficits characterizing neuropsychiatric disorders, including schizophrenia. Thus, there is growing interest in characterizing the relationship between cannabinoid transmission, emotional processing, and dopamine (DA)-dependent behavioral deficits. The CB1R is highly expressed in the mammalian nervous system, particularly in the hippocampus. Activation of the ventral hippocampal subregion (vHipp) is known to increase both the activity of DAergic neurons located in the ventral tegmental area (VTA) and DA levels in reward-related brain regions, particularly the nucleus accumbens (NAc). However, the possible functional relationship between hippocampal CB1R transmission and VTA DA neuronal activity is not currently understood. In this study, using in vivo neuronal recordings in rats, we demonstrate that activation of CB1R in the vHipp strongly increases VTA DA neuronal firing and bursting activity, while simultaneously decreasing the activity of VTA non-DA neurons. Furthermore, using a conditioned place preference procedure and a social interaction test, we report that intra-vHipp CB1R activation potentiates the reward salience of normally sub-threshold conditioning doses of opiates and induces deficits in natural sociability and social recognition behaviors. Finally, these behavioral effects were prevented by directly blocking NAc DAergic transmission. Collectively, these findings identify hippocampal CB1R transmission as a critical modulator of the mesolimbic DA pathway and in the processing of reward and social-related behavioral phenomena.


Subject(s)
Dopaminergic Neurons/physiology , Hippocampus/physiology , Interpersonal Relations , Receptor, Cannabinoid, CB1/metabolism , Reward , Ventral Tegmental Area/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Dopaminergic Neurons/drug effects , Hippocampus/drug effects , Male , Memory/drug effects , Memory/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Rats, Sprague-Dawley , Receptors, Dopamine , Spatial Behavior/drug effects , Spatial Behavior/physiology , Ventral Tegmental Area/drug effects
17.
J Neurosci ; 34(39): 13096-109, 2014 Sep 24.
Article in English | MEDLINE | ID: mdl-25253856

ABSTRACT

Disturbances in cortical cannabinoid CB1 receptor signaling are well established correlates of various neuropsychiatric disorders, including depression and schizophrenia. Importantly, the ability of cannabinoid transmission to modulate emotional processing is functionally linked to interactions with subcortical DA systems. While considerable evidence demonstrates that CB1 receptor-mediated modulation of emotional processing and related behaviors follows a biphasic functional curve, little is known regarding how CB1 signaling within cortical networks may interact with subcortical DAergic systems involved in emotional behavior regulation. Using a combination of in vivo electrophysiological recordings and behavioral pharmacology in rats, we investigated the relationship between mPFC cannabinoid transmission, fear memory formation, and subcortical DA neuron activity patterns. We report that direct intra-mPFC CB1 activation biphasically modulates spontaneous, subcortical VTA DA neuron activity in a dose-dependent fashion; while lower doses of a CB1 receptor agonist, WIN 55,212-2, significantly increased spontaneous firing and bursting rates of VTA DA neurons, higher doses strongly inhibited spontaneous DA neuron activity. Remarkably, this same dose-related functional difference was observed with the regulation of fear-related emotional memory formation. Thus, lower levels of CB1 activation potentiated the emotional salience of normally subthreshold fear memory, whereas higher levels completely blocked fear memory acquisition. Furthermore, while the potentiation of subthreshold fear memory salience was blocked by DA receptor antagonism, CB1-mediated blunting of suprathreshold fear memory was rescued by intra-VTA administration of a GABAB receptor antagonist, demonstrating that reversal of GABAergic inhibitory mechanisms in the VTA can reverse the inhibitory influence of intra-PFC CB1 transmission on mesolimbic DA activity.


Subject(s)
Fear , Memory , Prefrontal Cortex/physiology , Receptor, Cannabinoid, CB1/metabolism , Ventral Tegmental Area/physiology , Action Potentials , Animals , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Male , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Ventral Tegmental Area/cytology , Ventral Tegmental Area/metabolism
18.
Front Psychiatry ; 5: 73, 2014.
Article in English | MEDLINE | ID: mdl-25071606

ABSTRACT

Emerging evidence from both basic and clinical research demonstrates an important role for endocannabinoid (ECB) signaling in the processing of emotionally salient information, learning, and memory. Cannabinoid transmission within neural circuits involved in emotional processing has been shown to modulate the acquisition, recall, and extinction of emotionally salient memories and importantly, can strongly modulate the emotional salience of incoming sensory information. Two neural regions in particular, the medial prefrontal cortex (PFC) and the basolateral nucleus of the amygdala (BLA), play important roles in emotional regulation and contain high levels of cannabinoid receptors. Furthermore, both regions show profound abnormalities in neuropsychiatric disorders such as addiction and schizophrenia. Considerable evidence has demonstrated that cannabinoid transmission functionally interacts with dopamine (DA), a neurotransmitter system that is of exceptional importance for both addictive behaviors and the neuropsychopathology of disorders like schizophrenia. Research in our laboratory has focused on how cannabinoid transmission both within and extrinsic to the mesolimbic DA system, including the BLA → mPFC circuitry, can modulate both rewarding and aversive emotional information. In this review, we will summarize clinical and basic neuroscience research demonstrating the importance of cannabinoid signaling within this neural circuitry. In particular, evidence will be reviewed emphasizing the importance of cannabinoid signaling within the BLA → mPFC circuitry in the context of emotional salience processing, memory formation and memory-related plasticity. We propose that aberrant states of hyper or hypoactive ECB signaling within the amygdala-prefrontal cortical circuit may lead to dysregulation of mesocorticolimbic DA transmission controlling the processing of emotionally salient information. These disturbances may in turn lead to emotional processing, learning, and memory abnormalities related to various neuropsychiatric disorders, including addiction and schizophrenia-related psychoses.

19.
Prog Neurobiol ; 111: 34-52, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24025745

ABSTRACT

The reuniens and rhomboid nuclei, located in the ventral midline of the thalamus, have long been regarded as having non-specific effects on the cortex, while other evidence suggests that they influence behavior related to the photoperiod, hunger, stress or anxiety. We summarise the recent anatomical, electrophysiological and behavioral evidence that these nuclei also influence cognitive processes. The first part of this review describes the reciprocal connections of the reuniens and rhomboid nuclei with the medial prefrontal cortex and the hippocampus. The connectivity pattern among these structures is consistent with the idea that these ventral midline nuclei represent a nodal hub to influence prefrontal-hippocampal interactions. The second part describes the effects of a stimulation or blockade of the ventral midline thalamus on cortical and hippocampal electrophysiological activity. The final part summarizes recent literature supporting the emerging view that the reuniens and rhomboid nuclei may contribute to learning, memory consolidation and behavioral flexibility, in addition to general behavior and aspects of metabolism.


Subject(s)
Behavior/physiology , Electrophysiological Phenomena/physiology , Midline Thalamic Nuclei/anatomy & histology , Midline Thalamic Nuclei/physiology , Animals , Humans
20.
Neuropsychopharmacology ; 38(12): 2418-26, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23736315

ABSTRACT

The lateral habenula (LHb) is an epithalamic structure connected with both the basal ganglia and the limbic system and that exerts a major influence on midbrain monoaminergic nuclei. The current view is that LHb receives and processes cortical information in order to select proper strategies in a variety of behavior. Recent evidence indicates that LHb might also be implicated in hippocampus-dependent memory processes. However, if and how LHb functionally interacts with the dorsal hippocampus (dHPC) is still unknown. We therefore performed simultaneous recordings within LHb and dHPC in both anesthetized and freely moving rats. We first showed that a subset of LHb cells were phase-locked to hippocampal theta oscillations. Furthermore, LHb generated spontaneous theta oscillatory activity, which was highly coherent with hippocampal theta oscillations. Using reversible LHb inactivation, we found that LHb might regulate dHPC theta oscillations. In addition, we showed that LHb silencing altered performance in a hippocampus-dependent spatial recognition task. Finally, increased coherence between LHb and dHPC was positively correlated to the memory performance in this test. Collectively, these results suggest that LHb functionally interacts with the hippocampus and is involved in hippocampus-dependent spatial information processing.


Subject(s)
Habenula/physiology , Hippocampus/physiology , Memory/physiology , Neurons/physiology , Animals , Male , Rats , Rats, Long-Evans , Recognition, Psychology/physiology , Space Perception/physiology , Theta Rhythm/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...