Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 7545, 2023 Nov 20.
Article in English | MEDLINE | ID: mdl-37985770

ABSTRACT

Brain levels of the endocannabinoid 2-arachidonoylglycerol (2-AG) shape motivated behavior and nucleus accumbens (NAc) dopamine release. However, it is not clear whether mobilization of 2-AG specifically from midbrain dopamine neurons is necessary for dopaminergic responses to external stimuli predicting forthcoming reward. Here, we use a viral-genetic strategy to prevent the expression of the 2-AG-synthesizing enzyme diacylglycerol lipase α (DGLα) from ventral tegmental area (VTA) dopamine cells in adult mice. We find that DGLα deletion from VTA dopamine neurons prevents depolarization-induced suppression of excitation (DSE), a form of 2-AG-mediated synaptic plasticity, in dopamine neurons. DGLα deletion also decreases effortful, cue-driven reward-seeking but has no effect on non-cued or low-effort operant tasks and other behaviors. Moreover, dopamine recording in the NAc reveals that deletion of DGLα impairs the transfer of accumbal dopamine signaling from a reward to its earliest predictors. These results demonstrate that 2-AG mobilization from VTA dopamine neurons is a necessary step for the generation of dopamine-based predictive associations that are required to direct and energize reward-oriented behavior.


Subject(s)
Dopamine , Dopaminergic Neurons , Mice , Animals , Dopaminergic Neurons/metabolism , Dopamine/metabolism , Endocannabinoids/metabolism , Ventral Tegmental Area/physiology , Reward
2.
Cell Rep ; 42(6): 112553, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37224011

ABSTRACT

Understanding mesolimbic dopamine adaptations underlying vulnerability to drug relapse is essential to inform prognostic tools for effective treatment strategies. However, technical limitations have hindered the direct measurement of sub-second dopamine release in vivo for prolonged periods of time, making it difficult to gauge the weight that these dopamine abnormalities have in determining future relapse incidence. Here, we use the fluorescent sensor GrabDA to record, with millisecond resolution, every single cocaine-evoked dopamine transient in the nucleus accumbens (NAc) of freely moving mice during self-administration. We reveal low-dimensional features of patterned dopamine release that are strong predictors of cue-induced reinstatement of cocaine seeking. Additionally, we report sex-specific differences in cocaine-related dopamine responses related to a greater resistance to extinction in males compared with females. These findings provide important insights into the sufficiency of NAc dopamine signaling dynamics-in interaction with sex-for recapitulating persistent cocaine seeking and future relapse vulnerability.


Subject(s)
Cocaine-Related Disorders , Cocaine , Rats , Male , Mice , Animals , Cocaine/pharmacology , Dopamine/pharmacology , Rats, Sprague-Dawley , Conditioning, Operant , Extinction, Psychological/physiology , Recurrence , Nucleus Accumbens/physiology , Cues
3.
J Neurosci ; 43(25): 4684-4696, 2023 06 21.
Article in English | MEDLINE | ID: mdl-37208179

ABSTRACT

Sign-tracking (ST) rats show enhanced cue sensitivity before drug experience that predicts greater discrete cue-induced drug seeking compared with goal-tracking or intermediate rats. Cue-evoked dopamine in the nucleus accumbens (NAc) is a neurobiological signature of sign-tracking behaviors. Here, we examine a critical regulator of the dopamine system, endocannabinoids, which bind the cannabinoid receptor-1 (CB1R) in the ventral tegmental area (VTA) to control cue-evoked striatal dopamine levels. We use cell type-specific optogenetics, intra-VTA pharmacology, and fiber photometry to test the hypothesis that VTA CB1R receptor signaling regulates NAc dopamine levels to control sign tracking. We trained male and female rats in a Pavlovian lever autoshaping (PLA) task to determine their tracking groups before testing the effect of VTA → NAc dopamine inhibition. We found that this circuit is critical for mediating the vigor of the ST response. Upstream of this circuit, intra-VTA infusions of rimonabant, a CB1R inverse agonist, during PLA decrease lever and increase food cup approach in sign-trackers. Using fiber photometry to measure fluorescent signals from a dopamine sensor, GRABDA (AAV9-hSyn-DA2m), we tested the effects of intra-VTA rimonabant on NAc dopamine dynamics during autoshaping in female rats. We found that intra-VTA rimonabant decreased sign-tracking behaviors, which was associated with increases in NAc shell, but not core, dopamine levels during reward delivery [unconditioned stimulus (US)]. Our results suggest that CB1R signaling in the VTA influences the balance between the conditioned stimulus-evoked and US-evoked dopamine responses in the NAc shell and biases behavioral responding to cues in sign-tracking rats.SIGNIFICANCE STATEMENT Substance use disorder (SUD) is a chronically relapsing psychological disorder that affects a subset of individuals who engage in drug use. Recent research suggests that there are individual behavioral and neurobiological differences before drug experience that predict SUD and relapse vulnerabilities. Here, we investigate how midbrain endocannabinoids regulate a brain pathway that is exclusively involved in driving cue-motivated behaviors of sign-tracking rats. This work contributes to our mechanistic understanding of individual vulnerabilities to cue-triggered natural reward seeking that have relevance for drug-motivated behaviors.


Subject(s)
Nucleus Accumbens , Ventral Tegmental Area , Female , Rats , Male , Animals , Nucleus Accumbens/physiology , Ventral Tegmental Area/physiology , Cues , Dopamine/metabolism , Endocannabinoids/pharmacology , Rimonabant/pharmacology , Drug Inverse Agonism , Reward , Polyesters/metabolism , Polyesters/pharmacology
4.
Neuropsychopharmacology ; 48(12): 1821-1831, 2023 11.
Article in English | MEDLINE | ID: mdl-37208501

ABSTRACT

Impulsive choice has enduring trait-like characteristics and is defined by preference for small immediate rewards over larger delayed ones. Importantly, it is a determining factor in the development and persistence of substance use disorder (SUD). Emerging evidence from human and animal studies suggests frontal cortical regions exert influence over striatal reward processing areas during decision-making in impulsive choice or delay discounting (DD) tasks. The goal of this study was to examine how these circuits are involved in decision-making in animals with defined trait impulsivity. To this end, we trained adolescent male rats to stable behavior on a DD procedure and then re-trained them in adulthood to assess trait-like, conserved impulsive choice across development. We then used chemogenetic tools to selectively and reversibly target corticostriatal projections during performance of the DD task. The prelimbic region of the medial prefrontal cortex (mPFC) was injected with a viral vector expressing inhibitory designer receptors exclusively activated by designer drugs (Gi-DREADD), and then mPFC projections to the nucleus accumbens core (NAc) were selectively suppressed by intra-NAc administration of the Gi-DREADD actuator clozapine-n-oxide (CNO). Inactivation of the mPFC-NAc projection elicited a robust increase in impulsive choice in rats with lower vs. higher baseline impulsivity. This demonstrates a fundamental role for mPFC afferents to the NAc during choice impulsivity and suggests that maladaptive hypofrontality may underlie decreased executive control in animals with higher levels of choice impulsivity. Results such as these may have important implications for the pathophysiology and treatment of impulse control, SUDs, and related psychiatric disorders.


Subject(s)
Impulsive Behavior , Prefrontal Cortex , Adolescent , Rats , Male , Humans , Animals , Impulsive Behavior/physiology , Prefrontal Cortex/physiology , Reward , Nucleus Accumbens/physiology , Choice Behavior/physiology
5.
Exp Neurol ; 364: 114384, 2023 06.
Article in English | MEDLINE | ID: mdl-36934867

ABSTRACT

Over the past few decades, tremendous work has been dedicated to understanding how cannabinoids, both endogenous and exogenous, impact the process of learning and memory. Here we attempt to summarize the breadth of this research investigating the role of cannabinoid signaling on episodic or hippocampus-dependent memory. This review will focus primarily on studies using pharmacological approaches, allowing us to discuss the impact of cannabinoids at different phases of the memory formation process. We will briefly provide an overview of the endocannabinoid system and the mechanisms underlying memory formation, then discuss the impact of cannabinoids on memory encoding, consolidation, retrieval, and extinction. Additionally, we examine the impact of cannabinoids on synaptic plasticity and intracellular signaling within the hippocampus related to these processes.


Subject(s)
Cannabinoids , Endocannabinoids , Cannabinoids/pharmacology , Learning , Memory/physiology , Hippocampus
6.
Addict Neurosci ; 32022 Sep.
Article in English | MEDLINE | ID: mdl-36419922

ABSTRACT

Cannabinoid type 1 receptors (CB1Rs) orchestrate brain reward circuitry and are prevalent neurobiological targets for endocannabinoids and cannabis in the mammalian brain. Decades of histological and electrophysiological studies have established CB1R as presynaptic G-protein coupled receptors (GPCRs) that inhibit neurotransmitter release through retrograde signaling mechanisms. Recent seminal work demonstrates CB1R expression on astrocytes and the pivotal function of glial cells in endocannabinoid-mediated modulation of neuron-astrocyte signaling. Here, we review key facets of CB1R-mediated astroglia regulation of synaptic glutamate transmission in the nucleus accumbens with a specific emphasis on cocaine-directed behaviors.

7.
Nat Rev Neurosci ; 23(4): 191-203, 2022 04.
Article in English | MEDLINE | ID: mdl-35228740

ABSTRACT

Central nervous system neurons communicate via fast synaptic transmission mediated by ligand-gated ion channel (LGIC) receptors and slower neuromodulation mediated by G protein-coupled receptors (GPCRs). These receptors influence many neuronal functions, including presynaptic neurotransmitter release. Presynaptic LGIC and GPCR activation by locally released neurotransmitters influences neuronal communication in ways that modify effects of somatic action potentials. Although much is known about presynaptic receptors and their mechanisms of action, less is known about when and where these receptor actions alter release, especially in vivo. This Review focuses on emerging evidence for important local presynaptic receptor actions and ideas for future studies in this area.


Subject(s)
Cell Communication , Receptors, Presynaptic , Action Potentials , Humans , Neurons , Synaptic Transmission
8.
J Neurosci ; 41(32): 6946-6953, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34230105

ABSTRACT

Motivational deficits characterized by an unwillingness to overcome effortful costs are a common feature of neuropsychiatric and neurologic disorders that are insufficiently understood and treated. Dopamine (DA) signaling in the nucleus accumbens (NAc) facilitates goal-seeking, but how NAc DA release encodes motivationally salient stimuli to influence effortful investment is not clear. Using fast-scan cyclic voltammetry in male and female mice, we find that NAc DA release diametrically responds to cues signaling increasing cost of reward, while DA release to the reward itself is unaffected by its cost. Because endocannabinoid (eCB) signaling facilitates goal seeking and NAc DA release, we further investigated whether repeated augmentation of the eCB 2-arachidonoylglycerol with a low dose of a monoacylglycerol lipase (MAGL) inhibitor facilitates motivation and DA signaling without the development of tolerance. We find that chronic MAGL treatment stably facilitates goal seeking and DA encoding of prior reward cost, providing critical insight into the neurobiological mechanisms of a viable treatment for motivational deficits.SIGNIFICANCE STATEMENT Decades of work has established a fundamental role for dopamine neurotransmission in motivated behavior and cue-reward learning, but how dopaminergic encoding of cues associates with motivated action has remained unclear. Specifically, how dopamine neurons signal future and prior reward cost, and whether this can be modified to influence motivational set points is not known. The current study provides important insight into how dopamine neurons encode motivationally relevant stimuli to influence goal-directed action and supports cannabinoid-based therapies for treatment of motivational disorders.


Subject(s)
Dopaminergic Neurons/physiology , Endocannabinoids/metabolism , Motivation/physiology , Nucleus Accumbens/physiology , Reward , Animals , Cues , Dopamine , Dopaminergic Neurons/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Monoacylglycerol Lipases/pharmacology , Motivation/drug effects , Nucleus Accumbens/drug effects
9.
Neuropharmacology ; 193: 108625, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34058192

ABSTRACT

A wide body of evidence supports an integral role for mesolimbic dopamine (DA) in motivated behavior. In brief, drugs that increase DA in mesolimbic terminal regions, like cocaine, enhance motivation, while drugs that decrease DA concentration reduce motivation. Data from our laboratory and others shows that phasic activation of mesolimbic DA requires signaling at cannabinoid type-1 (CB1) receptors in the ventral tegmental area (VTA), and systemic delivery of CB1 receptor antagonists reduces DA cell activity and attenuates motivated behaviors. Recent findings demonstrate that cocaine mobilizes the endocannabinoid 2-arachidonoylglycerol (2-AG) in the VTA to cause phasic activation of DA neurons and terminal DA release. It remains unclear, however, if cocaine-induced midbrain 2-AG signaling contributes to the motivation-enhancing effects of cocaine. To examine this, we trained male and female rats on a progressive ratio (PR) task for a food reinforcer. Each rat underwent a series of tests in which they were pretreated with cocaine alone or in combination with systemic or intra-VTA administration of the CB1 receptor antagonist rimonabant or the 2-AG synthesis inhibitor tetrahydrolipstatin (THL). Cocaine increased motivation, measured by augmented PR breakpoints, while rimonabant dose-dependently decreased motivation. Importantly, intra-VTA administration of rimonabant or THL, at doses that did not decrease breakpoints on their own, blocked systemic cocaine administration from increasing breakpoints in male and female rats. These data suggest that cocaine-induced increases in motivation require 2-AG signaling at CB1 receptors in the VTA and may provide critical insight into cannabinoid-based pharmacotherapeutic targets for the successful treatment of substance abuse.


Subject(s)
Arachidonic Acids/antagonists & inhibitors , Cocaine/pharmacology , Endocannabinoids/antagonists & inhibitors , Glycerides/antagonists & inhibitors , Motivation/drug effects , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Ventral Tegmental Area/drug effects , Animals , Conditioning, Operant/drug effects , Female , Male , Rats , Rats, Long-Evans , Reward , Rimonabant/pharmacology , Self Administration
10.
Trends Neurosci ; 44(6): 464-477, 2021 06.
Article in English | MEDLINE | ID: mdl-33674134

ABSTRACT

Dopamine (DA), serotonin (5-hydroxytryptamine, 5-HT), and endocannabinoids (ECs) are key neuromodulators involved in many aspects of motivated behavior, including reward processing, reinforcement learning, and behavioral flexibility. Among the longstanding views about possible relationships between these neuromodulators is the idea of DA and 5-HT acting as opponents. This view has been challenged by emerging evidence that 5-HT supports reward seeking via activation of DA neurons in the ventral tegmental area. Adding an extra layer of complexity to these interactions, the endocannabinoid system is uniquely placed to influence dopaminergic and serotonergic neurotransmission. In this review we discuss how these three neuromodulatory systems interact at the cellular and circuit levels. Technological advances that facilitate precise identification and control of genetically targeted neuronal populations will help to achieve a better understanding of the complex relationship between these essential systems, and the potential relevance for motivated behavior.


Subject(s)
Endocannabinoids , Serotonin , Dopamine , Dopaminergic Neurons , Humans , Neurotransmitter Agents , Reward , Ventral Tegmental Area
11.
Neuropharmacology ; 189: 108527, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33741403

ABSTRACT

Binge ethanol drinking is an increasingly problematic component of alcohol use disorder costing the United States approximately over $150 billion every year and causes progressive neuroplasticity alterations in numerous brain regions. However, the precise nature or machinery that underlies binge drinking has not yet been elucidated. Corticotropin releasing factor (CRF) neurons in the central amygdala (CeA) are thought to modulate binge drinking, but the specific circuit mechanisms remain poorly understood. Here, we combined optogenetics with in vivo electrophysiology to identify and record from CeA CRF neurons in mice during a repeated binge ethanol drinking task. First, we found that CeA CRF neurons were more active than CeA non-CRF cells during our binge drinking paradigm. We also observed that CeA CRF neurons displayed a heterogeneous spectrum of responses to a lick of ethanol including, pre-lick activated, lick-excited, lick-inhibited, and no response. Interestingly, pre-lick activated CeA CRF neurons exhibited higher frequency and burst firing during binge drinking sessions. Moreover, their overall tonic and phasic electrical activity enhances over repeated binge drinking sessions. Remarkably, CeA CRF units and pre-lick activated CeA CRF neurons did not show higher firing rate or bursting activity during water and sucrose consumption, suggesting that ethanol may "hijack" or plastically alter their intrinsic excitability. This article is part of the special issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.


Subject(s)
Action Potentials/physiology , Binge Drinking/metabolism , Central Amygdaloid Nucleus/metabolism , Corticotropin-Releasing Hormone/metabolism , Ethanol/toxicity , Neurons/metabolism , Action Potentials/drug effects , Alcohol Drinking/adverse effects , Alcohol Drinking/physiopathology , Animals , Binge Drinking/physiopathology , Central Amygdaloid Nucleus/drug effects , Central Amygdaloid Nucleus/physiopathology , Ethanol/administration & dosage , Female , Male , Mice , Mice, Transgenic , Microelectrodes , Neurons/drug effects
12.
Addict Biol ; 26(3): e12961, 2021 05.
Article in English | MEDLINE | ID: mdl-32820590

ABSTRACT

Individuals suffering from substance use disorder often experience relapse events that are attributed to drug craving. Insular cortex (IC) function is implicated in processing drug-predictive cues and is thought to be a critical substrate for drug craving, but the downstream neural circuit effectors of the IC that mediate reward processing are poorly described. Here, we uncover the functional connectivity of an IC projection to the ventral bed nucleus of the stria terminalis (vBNST), a portion of the extended amygdala that has been previously shown to modulate dopaminergic activity within the ventral tegmental area (VTA), and investigate the role of this pathway in reward-related behaviors. We utilized ex vivo slice electrophysiology and in vivo optogenetics to examine the functional connectivity of the IC-vBNST projection and bidirectionally control IC-vBNST terminals in various reward-related behavioral paradigms. We hypothesized that the IC recruits mesolimbic dopamine signaling by activating VTA-projecting, vBNST neurons. Using slice electrophysiology, we found that the IC sends a glutamatergic projection onto vBNST-VTA neurons. Photoactivation of IC-vBNST terminals was sufficient to reinforce behavior in a dopamine-dependent manner. Moreover, silencing the IC-vBNST projection was aversive and resulted in anxiety-like behavior without affecting food consumption. This work provides a potential mechanism by which the IC processes exteroceptive triggers that are predictive of reward.


Subject(s)
Behavior, Animal/physiology , Cerebral Cortex/physiology , Dopamine/metabolism , Septal Nuclei/physiology , Amygdala/physiology , Animals , Anxiety/physiopathology , Female , Humans , Male , Mice, Inbred C57BL , Reward , Ventral Tegmental Area/physiology
13.
Article in English | MEDLINE | ID: mdl-31964646

ABSTRACT

Cannabis sativa, like all known drugs of abuse, leads to increased dopamine activation within the mesolimbic pathway. Consequent dopamine release within terminal regions of the striatum is a powerful mediator of reward and reinforcement and patterned dopamine release is critical for associative learning processes that are fundamentally involved in addiction. The endocannabinoid system modulates dopamine release at multiple sites, and the receptors, endogenous ligands, and synthetic and metabolic enzymes of the endocannabinoid system may provide key targets for pharmacotherapies to treat disorders of motivation including addiction. Disrupting endocannabinoid signaling decreases drug-induced increases in dopamine release as well those dopamine events evoked by conditioned stimuli during reward seeking. Advances in recording techniques for dopamine are allowing unprecedented examinations of these two interacting systems and elucidating the mechanisms of endocannabinoid modulation of dopamine release in reward and addiction.


Subject(s)
Brain/drug effects , Cannabinoid Receptor Agonists/pharmacology , Dopamine/metabolism , Marijuana Abuse/metabolism , Reward , Animals , Brain/physiology , Dronabinol/pharmacology , Endocannabinoids/physiology , Humans , Marijuana Abuse/physiopathology , Marijuana Abuse/psychology , Mice , Signal Transduction/drug effects , Signal Transduction/physiology , Substance Withdrawal Syndrome/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
14.
Curr Opin Pharmacol ; 56: 29-38, 2021 02.
Article in English | MEDLINE | ID: mdl-33068883

ABSTRACT

Prolonged exposure to drugs of abuse leads to severe alterations in mesocorticolimbic dopamine circuitry deeply implicated in substance use disorders. Despite considerable efforts, few medications to reduce relapse rates are currently available. To solve this issue, researchers are uncovering therapeutic opportunities offered by the endocannabinoid system. The cannabinoid receptor type 1 (CB1R), and its endogenous ligands, participate in orchestration of cue-triggered and stress-triggered responses leading to obtain natural and drug rewards. Here, we review the evidence supporting the use of CB1R neutral antagonists, allosteric modulators, indirect agonists, as well as multi-target compounds, as improved alternatives compared to classical CB1R antagonists. The promising therapeutic value of other substrates participating in endocannabinoid signaling, like peroxisome proliferator-activated receptors, is also covered. Overall, a wide body of pre-clinical evidence avails novel pharmacological strategies interacting with the endocannabinoid system as clinically amenable candidates able to counteract drug-induced dopamine maladaptations contributing to increased risk of relapse.


Subject(s)
Pharmaceutical Preparations , Substance-Related Disorders , Endocannabinoids , Humans , Ligands , Reward , Substance-Related Disorders/drug therapy , Substance-Related Disorders/prevention & control
15.
Article in English | MEDLINE | ID: mdl-33031862

ABSTRACT

Cannabis is the illicit drug most widely used by pregnant women worldwide. Its growing acceptance and legalization have markedly increased the risks of child psychopathology, including psychotic-like experiences, which lowers the age of onset for a first psychotic episode. As the majority of patients with schizophrenia go through a premorbid condition long before this occurs, understanding neurobiological underpinnings of the prodromal stage of the disease is critical to improving illness trajectories and therapeutic outcomes. We have previously shown that male rat offspring prenatally exposed to Δ9-tetrahydrocannabinol (THC), a rat model of prenatal cannabinoid exposure (PCE), exhibit extensive molecular and synaptic changes in dopaminergic neurons of the ventral tegmental area (VTA), converging on a hyperdopaminergic state. This leads to a silent psychotic-like endophenotype that is unmasked by a single exposure to THC. Here, we further characterized the VTA dopamine neuron and sensorimotor gating functions of PCE rats exposed to acute stress or a challenge of the D2 receptor agonist apomorphine, by using in vivo single-unit extracellular recordings and Prepulse Inhibition (PPI) analyses. At pre-puberty, PCE male rat offspring display a reduced population activity of VTA dopamine neurons in vivo, the majority of which are tonically active. PCE male progeny also exhibit enhanced sensitivity to dopamine D2 (DAD2) receptor activation and a vulnerability to acute stress, which is associated with compromised sensorimotor gating functions. This data extends our knowledge of the multifaceted sequelae imposed by PCE in the mesolimbic dopamine system of male pre-adolescent rats, which renders a neural substrate highly susceptible to subsequent challenges that may trigger psychotic-like outcomes.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dronabinol/pharmacology , Nucleus Accumbens/drug effects , Prenatal Exposure Delayed Effects/metabolism , Ventral Tegmental Area/drug effects , Animals , Dopaminergic Neurons/metabolism , Female , Male , Nucleus Accumbens/metabolism , Pregnancy , Prepulse Inhibition/drug effects , Rats , Rats, Sprague-Dawley , Reflex, Startle/drug effects , Ventral Tegmental Area/metabolism
16.
Neuropharmacology ; 182: 108374, 2021 01.
Article in English | MEDLINE | ID: mdl-33115642

ABSTRACT

Synthetic cannabinoids were introduced into recreational drug culture in 2008 and quickly became one of the most commonly abused drugs in the United States. The neurobiological consequences resulting from synthetic cannabinoid repeated exposure remain poorly understood. It is possible that a blunted dopamine (DA) response may lead drug users to consume larger quantities to compensate for this form of neurochemical tolerance. Because the endogenous cannabinoid and opioid systems exhibit considerable cross-talk and cross-tolerance frequently develops following repeated exposure to either opioids or cannabinoids, there is interest in investigating whether a history of synthetic cannabinoid exposure influences the ability of heroin to increase DA release. To test the effects of chronic cannabinoid exposure on cannabinoid- and heroin-evoked DA release, male adult rats were treated with either vehicle or a synthetic cannabinoid (WIN55-212-2; WIN) using an intravenous (IV) dose escalation regimen (0.2-0.8 mg/kg IV over 9 treatments). As predicted, WIN-treated rats showed a rightward shift in the dose-response relationship across all behavioral/physiological measures when compared to vehicle-treated controls. Then, using fast-scan cyclic voltammetry to measure changes in the frequency of transient DA events in the nucleus accumbens shell of awake and freely-moving rats, it was observed that the DA releasing effects of both WIN and heroin were significantly reduced in male rats with a pharmacological history of cannabinoid exposure. These results demonstrate that repeated exposure to the synthetic cannabinoid WIN can produce tolerance to its DA releasing effects and cross-tolerance to the DA releasing effects of heroin.


Subject(s)
Analgesics, Opioid/pharmacology , Benzoxazines/pharmacology , Cannabinoids/administration & dosage , Dopamine/metabolism , Drug Tolerance/physiology , Heroin/pharmacology , Morpholines/pharmacology , Naphthalenes/pharmacology , Age Factors , Analgesics , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Male , Pain Measurement/drug effects , Pain Measurement/methods , Rats , Rats, Long-Evans
17.
Curr Biol ; 30(19): 3724-3735.e2, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32763169

ABSTRACT

The ability to recognize emotions in others and adapt one's behavior accordingly is critical for functioning in any social context. This ability is impaired in several psychiatric disorders, such as autism and psychopathy. Recent work has identified the anterior cingulate cortex (ACC) among other brain regions involved in this process. Neural recording studies have shown that neurons in ACC are modulated by reward or shock when delivered to a conspecific and when experienced first-hand. Because previous studies do not vary reward and shock within the same experiment, it has been unclear whether the observed activity reflects how much attention is being paid to outcomes delivered to a conspecific or the valence associated with those stimuli. To address this issue, we recorded from ACC as rats performed a Pavlovian task that predicted whether reward, shock, or nothing would be delivered to the rat being recorded from or a conspecific located in the opposite chamber. Consistent with previous reports, we found that the firing of ACC neurons was modulated by aversive stimuli delivered to the recording rat and their conspecific. Activity of some of these neurons genuinely reflected outcome identity (i.e., reward or shock); however, the population of neurons as a whole responded similarly for both reward and shock, as well as for cues that predicted their occurrence (i.e., reward > neutral and shock > neutral; attention). These results suggest that ACC can process information about outcomes (i.e., identity and recipient) in the service of promoting attention in some social contexts.


Subject(s)
Empathy/physiology , Gyrus Cinguli/metabolism , Gyrus Cinguli/physiology , Action Potentials/physiology , Animals , Attention/physiology , Behavior, Animal/physiology , Brain/physiology , Choice Behavior/physiology , Cues , Electrophysiology/methods , Emotions , Male , Neurons/physiology , Rats , Rats, Sprague-Dawley , Reaction Time/physiology , Reward , Social Behavior
18.
Mol Psychiatry ; 25(11): 2786-2799, 2020 11.
Article in English | MEDLINE | ID: mdl-30116027

ABSTRACT

Recent clinical and preclinical studies suggest that selective activators of the M4 muscarinic acetylcholine receptor have potential as a novel treatment for schizophrenia. M4 activation inhibits striatal dopamine release by mobilizing endocannabinoids, providing a mechanism for local effects on dopamine signaling in the striatum but not in extrastriatal areas. G protein-coupled receptors (GPCRs) typically induce endocannabinoid release through activation of Gαq/11-type G proteins whereas M4 transduction occurs through Gαi/o-type G proteins. We now report that the ability of M4 to inhibit dopamine release and induce antipsychotic-like effects in animal models is dependent on co-activation of the Gαq/11-coupled mGlu1 subtype of metabotropic glutamate (mGlu) receptor. This is especially interesting in light of recent findings that multiple loss of function single nucleotide polymorphisms (SNPs) in the human gene encoding mGlu1 (GRM1) are associated with schizophrenia, and points to GRM1/mGlu1 as a gene within the "druggable genome" that could be targeted for the treatment of schizophrenia. Herein, we report that potentiation of mGlu1 signaling following thalamo-striatal stimulation is sufficient to inhibit striatal dopamine release, and that a novel mGlu1 positive allosteric modulator (PAM) exerts robust antipsychotic-like effects through an endocannabinoid-dependent mechanism. However, unlike M4, mGlu1 does not directly inhibit dopamine D1 receptor signaling and does not reduce motivational responding. Taken together, these findings highlight a novel mechanism of cross talk between mGlu1 and M4 and demonstrate that highly selective mGlu1 PAMs may provide a novel strategy for the treatment of positive symptoms associated with schizophrenia.


Subject(s)
Allosteric Regulation/drug effects , Antipsychotic Agents/metabolism , Receptor, Muscarinic M4/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Glutamic Acid/metabolism , Mice , Mice, Inbred C57BL
19.
Nat Neurosci ; 22(12): 1975-1985, 2019 12.
Article in English | MEDLINE | ID: mdl-31611707

ABSTRACT

The increased legal availability of cannabis has led to a common misconception that it is a safe natural remedy for, among others, pregnancy-related ailments such as morning sickness. Emerging clinical evidence, however, indicates that prenatal cannabis exposure (PCE) predisposes offspring to various neuropsychiatric disorders linked to aberrant dopaminergic function. Yet, our knowledge of how cannabis exposure affects the maturation of this neuromodulatory system remains limited. Here, we show that male, but not female, offspring of Δ9-tetrahydrocannabinol (THC)-exposed dams, a rat PCE model, exhibit extensive molecular and synaptic changes in dopaminergic neurons of the ventral tegmental area, including altered excitatory-to-inhibitory balance and switched polarity of long-term synaptic plasticity. The resulting hyperdopaminergic state leads to increased behavioral sensitivity to acute THC exposure during pre-adolescence. The neurosteroid pregnenolone, a US Food and Drug Administration (FDA) approved drug, rescues synaptic defects and normalizes dopaminergic activity and behavior in PCE offspring, thus suggesting a therapeutic approach for offspring exposed to cannabis during pregnancy.


Subject(s)
Dopaminergic Neurons/metabolism , Dronabinol/adverse effects , Dronabinol/pharmacology , Pregnenolone/pharmacology , Prenatal Exposure Delayed Effects/metabolism , Animals , Dopamine/metabolism , Dopaminergic Neurons/physiology , Dronabinol/antagonists & inhibitors , Endophenotypes , Female , Maze Learning/drug effects , Membrane Potentials/physiology , Motor Activity/drug effects , Neural Inhibition/physiology , Neuronal Plasticity/drug effects , Nucleus Accumbens/metabolism , Pregnancy , Prepulse Inhibition/drug effects , Prepulse Inhibition/physiology , Rats , Risk-Taking , Sensory Gating/drug effects , Sensory Gating/physiology , Sex Characteristics , Ventral Tegmental Area/metabolism
20.
Neuropsychopharmacology ; 44(11): 1876-1885, 2019 10.
Article in English | MEDLINE | ID: mdl-31132785

ABSTRACT

Enkephalins, endogenous ligands for delta opioid receptors (DORs), are highly enriched in the nucleus accumbens (NAc). They are implicated in depression but their role in the NAc, a critical brain region for motivated behavior, is not fully investigated. To provide insight into enkephalin function we used a chronic social defeat stress paradigm, where animals are either categorized as susceptible or resilient to stress based on their performance in a social interaction test. Compared to controls, susceptible animals showed reduced enkephalin levels in the NAc. Such decrease in enkephalin levels is not due to a change in mRNA of its precursor protein, proenkephalin, in susceptible mice but is consistent with increased mRNA levels of enkephalinases in the NAc of susceptible animals. Systemic administration of enkephalinase inhibitors or NAc infusion of the DOR agonist, SNC80, caused a resilient outcome to CSDS. Both treatments increased phosphorylation of ERK, which was downregulated by social defeat stress. To further validate these results, we also used Q175 knock-in mice, an animal model of Huntington's disease in which enkephalin levels are reduced in striatum and comorbidity with mood disorders is common. Consistent with data in wild-type mice, Q175 animals showed reduced enkephalin levels in the NAc and enhanced susceptibility to a social defeat stress. Overall, our data implicate that depression-like behavior induced by social defeat stress arises from disrupted DOR signaling resulting from lowered levels of enkephalins, which is partly mediated through elevated expression of enkephalinases.


Subject(s)
Behavior, Animal/physiology , Dominance-Subordination , Enkephalins/metabolism , Nucleus Accumbens/metabolism , Stress, Psychological/metabolism , Analgesics, Opioid/pharmacology , Animals , Behavior, Animal/drug effects , Benzamides/pharmacology , Depression/metabolism , Disease Models, Animal , Mice , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Piperazines/pharmacology , Receptors, Opioid, delta/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...