Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.504
Filter
1.
Addict Biol ; 29(8): e13428, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39087789

ABSTRACT

The increasing rates of drug misuse highlight the urgency of identifying improved therapeutics for treatment. Most drug-seeking behaviours that can be modelled in rodents utilize the repeated intravenous self-administration (SA) of drugs. Recent studies examining the mesolimbic pathway suggest that Kv7/KCNQ channels may contribute to the transition from recreational to chronic drug use. However, to date, all such studies used noncontingent, experimenter-delivered drug model systems, and the extent to which this effect generalizes to rats trained to self-administer drugs is not known. Here, we tested the ability of retigabine (ezogabine), a Kv7 channel opener, to regulate instrumental behaviour in male Sprague Dawley rats. We first validated the ability of retigabine to target experimenter-delivered cocaine in a conditioned place preference (CPP) assay and found that retigabine reduced the acquisition of place preference. Next, we trained rats for cocaine-SA under a fixed-ratio or progressive-ratio reinforcement schedule and found that retigabine pretreatment attenuated the SA of low to moderate doses of cocaine. This was not observed in parallel experiments, with rats self-administering sucrose, a natural reward. Compared with sucrose-SA, cocaine-SA was associated with reductions in the expression of the Kv7.5 subunit in the nucleus accumbens, without alterations in Kv7.2 and Kv7.3. Therefore, these studies reveal a reward-specific reduction in SA behaviour and support the notion that Kv7 is a potential therapeutic target for human psychiatric diseases with dysfunctional reward circuitry.


Subject(s)
Carbamates , Cocaine , Phenylenediamines , Rats, Sprague-Dawley , Self Administration , Sucrose , Animals , Phenylenediamines/pharmacology , Phenylenediamines/administration & dosage , Carbamates/pharmacology , Carbamates/administration & dosage , Cocaine/pharmacology , Cocaine/administration & dosage , Male , Rats , Sucrose/administration & dosage , Sucrose/pharmacology , Drug-Seeking Behavior/drug effects , KCNQ Potassium Channels/drug effects , Conditioning, Operant/drug effects , Dopamine Uptake Inhibitors/pharmacology , Dopamine Uptake Inhibitors/administration & dosage
2.
J Neurosci Res ; 102(7): e25369, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39037062

ABSTRACT

Cannabis consumption has increased from 1.5% to 2.5% in Canada between 2012 and 2019. Clinical studies have indicated effects of prenatal cannabis exposure on birth weight, substance use, and neurodevelopmental disorders, but are confounded by several difficult to control variables. Animal models allow for examination of the mechanism of cannabis-induced changes in neurodevelopment and behavior, while controlling dose and timing. Several animal models of prenatal cannabis exposure exist which provide varying levels of construct validity, control of dose, and exposure to maternal stress. Using a voluntary oral consumption model, mouse dams received 5 mg/kg Δ9-tetrahydrocannabinol (THC) whole cannabis oil in peanut butter daily from gestational day 1 (GD1) to postnatal day 10 (PD10). At GD1, GD18, PD1, PD10, and PD15, maternal plasma was collected; pup brains were collected from GD18 onward. Pup brains had higher levels of THC and cannabidiol at each time point, each of which persisted in maternal plasma and pup brains past the end of treatment (PD15). Male and female adolescent offspring were examined for changes to ventral tegmental area (VTA) dopamine neuron activity and cocaine-seeking behavior. Prenatal and early postnatal (GD1-PD10) cannabis-exposed male, but not female mice had decreased gamma-aminobutyric acid (GABAergic) input, depolarized resting membrane potential, and increased spontaneous firing of VTA dopamine neurons. Cannabis-exposed offspring showed faster decay of N-methyl-D-aspartate (NMDA) currents in both sexes. However, no differences in cocaine-seeking behavior were noted. These data characterize a voluntary prenatal cannabis exposure model and demonstrates VTA dopamine neuronal activity is disinhibited in offspring.


Subject(s)
Cocaine , Dopaminergic Neurons , Prenatal Exposure Delayed Effects , Ventral Tegmental Area , Animals , Female , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , Pregnancy , Mice , Prenatal Exposure Delayed Effects/chemically induced , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Male , Cocaine/pharmacology , Cocaine/toxicity , Dronabinol/toxicity , Dronabinol/pharmacology , Mice, Inbred C57BL , Cannabis
3.
Transl Psychiatry ; 14(1): 269, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956048

ABSTRACT

Addiction is a complex behavioral disorder characterized by compulsive drug-seeking and drug use despite harmful consequences. The prefrontal cortex (PFC) plays a crucial role in cocaine addiction, involving decision-making, impulse control, memory, and emotional regulation. The PFC interacts with the brain's reward system, including the ventral tegmental area (VTA) and nucleus accumbens (NAc). The PFC also projects to the lateral habenula (LHb), a brain region critical for encoding negative reward and regulating the reward system. In the current study, we examined the role of PFC-LHb projections in regulating cocaine reward-related behaviors. We found that optogenetic stimulation of the PFC-LHb circuit during cocaine conditioning abolished cocaine preference without causing aversion. In addition, increased c-fos expression in LHb neurons was observed in animals that received optic stimulation during cocaine conditioning, supporting the circuit's involvement in cocaine preference regulation. Molecular analysis in animals that received optic stimulation revealed that cocaine-induced alterations in the expression of GluA1 subunit of AMPA receptor was normalized to saline levels in a region-specific manner. Moreover, GluA1 serine phosphorylation on S845 and S831 were differentially altered in LHb and VTA but not in the PFC. Together these findings highlight the critical role of the PFC-LHb circuit in controlling cocaine reward-related behaviors and shed light on the underlying mechanisms. Understanding this circuit's function may provide valuable insights into addiction and contribute to developing targeted treatments for substance use disorders.


Subject(s)
Cocaine , Habenula , Neurons , Optogenetics , Prefrontal Cortex , Receptors, AMPA , Reward , Animals , Prefrontal Cortex/metabolism , Cocaine/pharmacology , Male , Habenula/metabolism , Neurons/metabolism , Receptors, AMPA/metabolism , Cocaine-Related Disorders/physiopathology , Cocaine-Related Disorders/metabolism , Neural Pathways , Rats , Proto-Oncogene Proteins c-fos/metabolism , Phosphorylation , Ventral Tegmental Area/metabolism , Behavior, Animal
4.
Article in English | MEDLINE | ID: mdl-38950842

ABSTRACT

Acute stimulation of M1 or M4 muscarinic cholinergic receptors reduces cocaine abuse-related effects in mice and rats. The combined activation of these receptor subtypes produces synergistic effects on some behavioural endpoints in mice. M1 and M1 + M4 receptor stimulation in a cocaine vs. food choice assay in rats and microdialysis in rats showed delayed and lasting "anticocaine effects". Here, we tested whether these putative lasting neuroplastic changes are sufficient to occlude the reinforcing effects of cocaine at the behavioural level in mice. Mice were pre-treated with the M1 receptor partial agonist VU0364572, M4 receptor positive allosteric modulator VU0152100, or VU0364572 + VU0152100 two weeks prior to acquisition of cocaine intravenous self-administration (IVSA). Male C57BL/6JRj mice received vehicle, VU0364572, VU0152100, or VU0364572 + VU0152100. Female mice were tested with two VU0364572 + VU0152100 dose combinations or vehicle. To attribute potential effects to either reduced rewarding effects or increased aversion to cocaine, we tested VU0364572 alone and VU0364572 + VU0152100 in acquisition of cocaine-conditioned place preference (CPP) in male mice using an unbiased design. The acquisition of cocaine IVSA was drastically reduced and/or slowed in male and female mice receiving VU0364572 + VU0152100, but not either drug alone. Food-maintained operant behaviour was unaffected, indicating that the treatment effects were cocaine-specific. No treatment altered the acquisition of cocaine-CPP, neither in the post-test, nor in a challenge 14 days later. The cocaine IVSA findings confirm unusual long-lasting "anticocaine" effects of muscarinic M1 + M4 receptor stimulation. Thus, in mice, simultaneous stimulation of both receptor subtypes seems to produce potential neuroplastic changes that yield lasting effects.


Subject(s)
Cocaine , Mice, Inbred C57BL , Receptor, Muscarinic M1 , Receptor, Muscarinic M4 , Reinforcement, Psychology , Self Administration , Animals , Male , Cocaine/pharmacology , Cocaine/administration & dosage , Female , Receptor, Muscarinic M4/metabolism , Receptor, Muscarinic M1/agonists , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M1/drug effects , Mice , Dopamine Uptake Inhibitors/pharmacology , Dopamine Uptake Inhibitors/administration & dosage , Muscarinic Agonists/pharmacology , Conditioning, Operant/drug effects
5.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39063123

ABSTRACT

Prolonged exposure to HIV-1 transactivator of transcription (Tat) protein dysregulates monoamine transmission, a physiological change implicated as a key factor in promoting neurocognitive disorders among people living with HIV. We have demonstrated that in vivo expression of Tat in Tat transgenic mice decreases dopamine uptake through both dopamine transporter (DAT) and norepinephrine transporter (NET) in the prefrontal cortex. Further, our novel allosteric inhibitor of monoamine transporters, SRI-32743, has been shown to attenuate Tat-inhibited dopamine transport through DAT and alleviates Tat-potentiated cognitive impairments. The current study reports the pharmacological profiles of SRI-32743 in basal and Tat-induced inhibition of human NET (hNET) function. SRI-32743 exhibited less affinity for hNET binding than desipramine, a classical NET inhibitor, but displayed similar potency for inhibiting hDAT and hNET activity. SRI-32743 concentration-dependently increased hNET affinity for [3H]DA uptake but preserved the Vmax of dopamine transport. SRI-32743 slowed the cocaine-mediated dissociation of [3H]Nisoxetine binding and reduced both [3H]DA and [3H]MPP+ efflux but did not affect d-amphetamine-mediated [3H]DA release through hNET. Finally, we determined that SRI-32743 attenuated a recombinant Tat1-86-induced decrease in [3H]DA uptake via hNET. Our findings demonstrated that SRI-32743 allosterically disrupts the recombinant Tat1-86-hNET interaction, suggesting a potential treatment for HIV-infected individuals with concurrent cocaine abuse.


Subject(s)
Cocaine , Norepinephrine Plasma Membrane Transport Proteins , tat Gene Products, Human Immunodeficiency Virus , Norepinephrine Plasma Membrane Transport Proteins/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/chemistry , Cocaine/pharmacology , Cocaine/metabolism , Humans , HIV-1/metabolism , HIV-1/drug effects , Quinazolines/pharmacology , Quinazolines/chemistry , Animals , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Protein Binding , Mice
6.
Neurobiol Learn Mem ; 213: 107961, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39025429

ABSTRACT

In an animal model of compulsive drug use, a subset of rats continues to self-administer cocaine despite footshock consequences and is considered punishment resistant. We recently found that punishment resistance is associated with habits that persist under conditions that typically encourage a transition to goal-directed control. Given that random ratio (RR) and random interval (RI) schedules of reinforcement influence whether responding is goal-directed or habitual, we investigated the influence of these schedules on punishment resistance for cocaine or food. Male and female Sprague Dawley rats were trained to self-administer either intravenous cocaine or food pellets on a seeking-taking chained schedule of reinforcement, with the seeking lever requiring completion of either an RR20 or RI60 schedule. Rats were then given four days of punishment testing with footshock administered at the completion of seeking on a random one-third of trials. For cocaine-trained rats, the RI60 schedule led to greater punishment resistance (i.e., more trials completed) than the RR20 schedule in males and females. For food-trained rats, the RI60 schedule led to greater punishment resistance (i.e., higher reward rates) than the RR20 schedule in female rats, although male rats showed punishment resistance on both RR20 and RI60 schedules. For both cocaine and food, we found that seeking responses were suppressed to a greater degree than reward rate with the RI60 schedule, whereas response rate and reward rate were equally suppressed with the RR20 schedule. This dissociation between punishment effects on reward rate and response rate with the RI60 schedule can be explained by the nonlinear relation between these variables on RI schedules, but it does not account for the enhanced resistance to punishment. Overall, the results show greater punishment resistance with the RI60 schedule as compared to the RR20 schedule, indicating that schedules of reinforcement are an influencing factor on resistance to negative consequences.


Subject(s)
Cocaine , Punishment , Rats, Sprague-Dawley , Reinforcement Schedule , Self Administration , Animals , Male , Female , Cocaine/administration & dosage , Cocaine/pharmacology , Rats , Conditioning, Operant/drug effects , Reinforcement, Psychology , Drug-Seeking Behavior/drug effects , Drug-Seeking Behavior/physiology
7.
ACS Chem Neurosci ; 15(12): 2420-2431, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38838000

ABSTRACT

Recently, the abuse of synthetic cathinones is increasing among young people. α-Pyrrolidinobutiothiophenone (α-PBT), a synthetic cathinone, is a designer drug that is freely traded online with no legal restrictions. Moreover, there is currently no scientific basis for legal regulation. Here, we examined the addictive properties of α-PBT using a drug discrimination (DD) task. We also investigated the role of α-PBT in brain stimulation reward (BSR) using an intracranial self-stimulation (ICSS) paradigm in rats. Initially, the rats were trained to discriminate between cocaine and saline. After the discrimination training criteria were met, we determined the dose-effect curves of cocaine and conducted generalization tests with α-PBT and α-pyrrolidinopentiothiophenone (α-PVT) using a cumulative dosing protocol. In a separate set of studies, we examined the dopaminergic mechanisms underlying the function of α-PBT as an interoceptive stimulus (17.8 mg/kg) by intraperitoneally injecting either the dopamine (DA) D1 antagonist SCH23390 (0.06 and 0.12 mg/kg) or the D2 antagonist eticlopride (0.05 and 0.1 mg/kg) 15 min before DD testing. Brain reward function was measured using an ICSS procedure to examine the effects of α-PBT on ICSS threshold under the frequency-rate procedure. Our results showed that α-PBT functioned as a discriminative cue similar to cocaine in rats. More importantly, SCH23390 abolished the effects of α-PBT as an interoceptive stimulus in a dose-dependent manner in rats trained to press a lever to receive cocaine. Similarly, eticlopride dose-dependently attenuated the effect of α-PBT used as a discriminative cue. Additionally, cumulative α-PBT administration dose-dependently lowered ICSS thresholds compared with those in saline-treated rats. Furthermore, α-PBT-induced potentiation of BSR was abolished by pretreatment with both SCH23390 and eticlopride. Taken together, our results suggest that α-PBT can function as a cocaine-like discriminative cue via the activation of D1 and D2 receptors. α-PBT also appears to influence BSR by reducing the brain reward threshold via changes in D1 and D2 receptors. The present study suggests that α-PBT could have addictive properties through DA D1 and D2 receptors and thus poses a threat to humans.


Subject(s)
Cocaine , Self Stimulation , Animals , Male , Self Stimulation/drug effects , Rats , Cocaine/pharmacology , Rats, Sprague-Dawley , Pyrrolidines/pharmacology , Reward , Dose-Response Relationship, Drug , Thiophenes/pharmacology , Benzazepines/pharmacology , Designer Drugs/pharmacology , Discrimination, Psychological/drug effects , Brain/drug effects , Brain/metabolism
8.
Proc Natl Acad Sci U S A ; 121(24): e2401929121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38843183

ABSTRACT

Punishment such as electric shock or physical discipline employs a mixture of physical pain and emotional distress to induce behavior modification. However, a neural circuit that produces behavior modification by selectively focusing the emotional component, while bypassing the pain typically induced by peripheral nociceptor activation, is not well studied. Here, we show that genetically silencing the activity of neurons expressing calcitonin gene-related peptide (CGRP) in the parabrachial nucleus blocks the suppression of addictive-like behavior induced by footshock. Furthermore, activating CGRP neurons suppresses not only addictive behavior induced by self-stimulating dopamine neurons but also behavior resulting from self-administering cocaine, without eliciting nocifensive reactions. Moreover, among multiple downstream targets of CGRP neurons, terminal activation of CGRP in the central amygdala is effective, mimicking the results of cell body stimulation. Our results indicate that unlike conventional electric footshock, stimulation of CGRP neurons does not activate peripheral nociceptors but effectively curb addictive behavior.


Subject(s)
Behavior, Addictive , Calcitonin Gene-Related Peptide , Neurons , Parabrachial Nucleus , Animals , Parabrachial Nucleus/metabolism , Parabrachial Nucleus/physiology , Calcitonin Gene-Related Peptide/metabolism , Mice , Neurons/metabolism , Neurons/physiology , Behavior, Addictive/metabolism , Male , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Cocaine/pharmacology , Behavior, Animal/physiology
9.
Physiol Behav ; 282: 114599, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38823754

ABSTRACT

Cocaine addiction is the third largest cause of overdose-related deaths in the United States. Research investigating therapeutic targets for cocaine reward processes is key to combating this issue. The neuropeptide oxytocin (OXT) has been shown to reduce cocaine reward processes, though specific mechanisms are not understood. This study examines the effect of intra-dorsal hippocampal (DH) OXT on the expression of cocaine context associations using a conditioned place preference (CPP) paradigm. In this paradigm, one of two visually distinct chambers is paired with a drug. With repeated pairings, control animals display preference for the drug-associated context by spending more time in that context at test. In the present study, four conditioning days took place where male and female rats were injected with either cocaine or saline and placed into the corresponding chamber. On test day, rats received infusions of OXT or saline (VEH) into the DH and were allowed access to both chambers. The results show that while VEH-infused rats displayed cocaine CPP, OXT-infused rats did not prefer the cocaine-paired chamber. These findings implicate the DH as necessary in the mechanism by which OXT acts to block the expression of cocaine-context associations, providing insight into how OXT may exert its therapeutic effect in cocaine reward processes.


Subject(s)
Cocaine , Hippocampus , Oxytocin , Animals , Oxytocin/pharmacology , Cocaine/pharmacology , Male , Female , Hippocampus/drug effects , Hippocampus/metabolism , Rats , Dopamine Uptake Inhibitors/pharmacology , Rats, Sprague-Dawley , Reward
10.
Article in English | MEDLINE | ID: mdl-38844126

ABSTRACT

Abuse-related drug usage is a public health issue. Drosophila melanogaster has been used as an animal model to study the biological effects of these psychoactive substances in preclinical studies. Our objective in this review is to evaluate the adverse effects produced by cocaine, nicotine, and marijuana during the development of D. melanogaster. We searched experimental studies in which D. melanogaster was exposed to these three psychoactive drugs in seven online databases up to January 2023. Two reviewers independently extracted the data. Fifty-one studies met eligibility criteria and were included in the data extraction: nicotine (n = 26), cocaine (n = 20), and marijuana (n = 5). Fifteen studies were eligible for meta-analysis. Low doses (∼0.6 mM) of nicotine increased locomotor activity in fruit flies, while high doses (≥3 mM) led to a decrease. Similarly, exposure to cocaine increased locomotor activity, resulting in decreased climbing response in D. melanogaster. Studies with exposure to marijuana did not present a profile for our meta-analysis. However, this drug has been less associated with locomotor changes, but alterations in body weight and fat content and changes in cardiac function. Our analyses have shown that fruit flies exposed to drugs of abuse during different developmental stages, such as larvae and adults, exhibit molecular, morphological, behavioral, and survival changes that are dependent on the dosage. These phenotypes resemble the adverse effects of psychoactive substances in clinical medicine.


Subject(s)
Cocaine , Drosophila melanogaster , Nicotine , Animals , Drosophila melanogaster/drug effects , Cocaine/pharmacology , Cocaine/adverse effects , Nicotine/pharmacology , Nicotine/adverse effects , Locomotion/drug effects , Cannabis/adverse effects
11.
Dev Psychobiol ; 66(6): e22514, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38922890

ABSTRACT

Repeated exposure to abused drugs leads to reorganizing synaptic connections in the brain, playing a pivotal role in the relapse process. Additionally, recent research has highlighted the impact of parental drug exposure before gestation on subsequent generations. This study aimed to explore the influence of parental morphine exposure 10 days prior to pregnancy on drug-induced locomotor sensitization. Adult male and female Wistar rats were categorized into morphine-exposed and control groups. Ten days after their last treatment, they were mated, and their male offspring underwent morphine, methamphetamine, cocaine, and nicotine-induced locomotor sensitization tests. The results indicated increased locomotor activity in both groups after drug exposure, although the changes were attenuated in morphine and cocaine sensitization among the offspring of morphine-exposed parents (MEPs). Western blotting analysis revealed altered levels of D2 dopamine receptors (D2DRs) in the prefrontal cortex and nucleus accumbens of the offspring from MEPs. Remarkably, despite not having direct in utero drug exposure, these offspring exhibited molecular alterations affecting morphine and cocaine-induced sensitization. The diminished sensitization to morphine and cocaine suggested the development of a tolerance phenotype in these offspring. The changes in D2DR levels in the brain might play a role in these adaptations.


Subject(s)
Cocaine , Locomotion , Morphine , Nucleus Accumbens , Prefrontal Cortex , Prenatal Exposure Delayed Effects , Rats, Wistar , Receptors, Dopamine D2 , Animals , Female , Morphine/pharmacology , Morphine/administration & dosage , Male , Cocaine/pharmacology , Cocaine/administration & dosage , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Prenatal Exposure Delayed Effects/chemically induced , Rats , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D2/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Locomotion/drug effects , Behavior, Animal/drug effects , Behavior, Animal/physiology , Narcotics/pharmacology , Paternal Exposure/adverse effects , Dopamine Uptake Inhibitors/pharmacology , Dopamine Uptake Inhibitors/administration & dosage , Motor Activity/drug effects , Motor Activity/physiology
12.
Behav Brain Res ; 471: 115097, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38878971

ABSTRACT

Neuroadaptive changes in the hippocampus underlie addictive-like behaviors in humans or animals chronically exposed to cocaine. miR-181a, which is widely expressed in the hippocampus, acts as a regulator for synaptic plasticity, while its role in drug reinstatement is unclear. In this study, we found that miR-181a regulates the reinstatement of cocaine conditioned place preference(CPP), and altered miR-181a expression changes the complexity of hippocampal neurons and the density and morphology of dendritic spines. By using a luciferase gene reporter, we found that miR-181a targets PRKAA1, an upstream molecule in the mTOR pathway. High miR-181a expression reduced the expression of the PRKAA1 mRNA and promoted mTOR activity and the reinstatement of cocaine CPP. These results indicate that miR-181a is involved in neuronal structural plasticity induced by reinstatement of cocaine CPP, possibly through the activation of the mTOR signaling pathway. This study provides new microRNA targets and a theoretical foundation for the prevention of cocaine-induced reinstatement.


Subject(s)
Cocaine , Hippocampus , MicroRNAs , TOR Serine-Threonine Kinases , MicroRNAs/metabolism , Animals , Hippocampus/metabolism , Hippocampus/drug effects , Cocaine/pharmacology , Male , TOR Serine-Threonine Kinases/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , AMP-Activated Protein Kinases/metabolism , Neurons/drug effects , Neurons/metabolism , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Dopamine Uptake Inhibitors/pharmacology , Cocaine-Related Disorders/metabolism , Mice , Rats , Rats, Sprague-Dawley
13.
Addict Biol ; 29(5): e13397, 2024 05.
Article in English | MEDLINE | ID: mdl-38711205

ABSTRACT

Neuronal ensembles in the medial prefrontal cortex mediate cocaine self-administration via projections to the nucleus accumbens. We have recently shown that neuronal ensembles in the prelimbic cortex form rapidly to mediate cocaine self-administration. However, the role of neuronal ensembles within the nucleus accumbens in initial cocaine-seeking behaviour remains unknown. Here, we sought to expand the current literature by testing the necessity of the cocaine self-administration ensemble in the nucleus accumbens core (NAcCore) 1 day after male and female rats acquire cocaine self-administration by using the Daun02 inactivation procedure. We found that disrupting the NAcCore ensembles after a no-cocaine reward-seeking test increased subsequent cocaine seeking, while disrupting NAcCore ensembles following a cocaine self-administration session decreased subsequent cocaine seeking. We then characterized neuronal cell type in the NAcCore using RNAscope in situ hybridization. In the no-cocaine session, we saw reduced dopamine D1 type neuronal activation, while in the cocaine self-administration session, we found preferential dopamine D1 type neuronal activity in the NAcCore.


Subject(s)
Cocaine , Drug-Seeking Behavior , Neurons , Nucleus Accumbens , Self Administration , Animals , Nucleus Accumbens/drug effects , Cocaine/pharmacology , Male , Female , Rats , Drug-Seeking Behavior/drug effects , Neurons/drug effects , Reward , Dopamine Uptake Inhibitors/pharmacology , Reinforcement, Psychology , Receptors, Dopamine D1 , Cocaine-Related Disorders/physiopathology , Rats, Sprague-Dawley , Prefrontal Cortex/drug effects
14.
J Neurosci ; 44(23)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38719446

ABSTRACT

Drugs of abuse cause changes in the prefrontal cortex (PFC) and associated regions that impair inhibitory control over drug-seeking. Breaking the contingencies between drug-associated cues and the delivery of the reward during extinction learning reduces relapse. Vagus nerve stimulation (VNS) has previously been shown to enhance extinction learning and reduce drug-seeking. Here we determined the effects of VNS-mediated release of brain-derived neurotrophic factor (BDNF) on extinction and cue-induced reinstatement in male rats trained to self-administer cocaine. Pairing 10 d of extinction training with VNS facilitated extinction and reduced drug-seeking behavior during reinstatement. Rats that received a single extinction session with VNS showed elevated BDNF levels in the medial PFC as determined via an enzyme-linked immunosorbent assay. Systemic blockade of tropomyosin receptor kinase B (TrkB) receptors during extinction, via the TrkB antagonist ANA-12, decreased the effects of VNS on extinction and reinstatement. Whole-cell recordings in brain slices showed that cocaine self-administration induced alterations in the ratio of AMPA and NMDA receptor-mediated currents in Layer 5 pyramidal neurons of the infralimbic cortex (IL). Pairing extinction with VNS reversed cocaine-induced changes in glutamatergic transmission by enhancing AMPAR currents, and this effect was blocked by ANA-12. Our study suggests that VNS consolidates the extinction of drug-seeking behavior by reversing drug-induced changes in synaptic AMPA receptors in the IL, and this effect is abolished by blocking TrkB receptors during extinction, highlighting a potential mechanism for the therapeutic effects of VNS in addiction.


Subject(s)
Drug-Seeking Behavior , Extinction, Psychological , Neuronal Plasticity , Prefrontal Cortex , Rats, Sprague-Dawley , Receptor, trkB , Vagus Nerve Stimulation , Animals , Male , Rats , Vagus Nerve Stimulation/methods , Drug-Seeking Behavior/physiology , Drug-Seeking Behavior/drug effects , Receptor, trkB/metabolism , Receptor, trkB/antagonists & inhibitors , Neuronal Plasticity/physiology , Neuronal Plasticity/drug effects , Extinction, Psychological/physiology , Extinction, Psychological/drug effects , Prefrontal Cortex/physiology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Self Administration , Cocaine/pharmacology , Cocaine/administration & dosage
15.
Eur Addict Res ; 30(3): 163-180, 2024.
Article in English | MEDLINE | ID: mdl-38710170

ABSTRACT

INTRODUCTION: The development of cocaine use disorder in females is suggested to be more strongly related to neural mechanisms underlying stress-reactivity, whereas in males it is suggested to be more strongly related to neural mechanisms underlying drug cue-reactivity. Existing evidence, however, is based on neuroimaging studies that either lack a control group and/or have very small sample sizes that do not allow to investigate sex differences. METHODS: The main objective of the current study was to investigate sex differences in the neural correlates of cocaine and negative emotional cue-reactivity within high-risk intranasal cocaine users (CUs: 31 males and 26 females) and non-cocaine-using controls (non-CUs: 28 males and 26 females). A region of interest (ROI) analysis was applied to test for the main and interaction effects of group, sex, and stimulus type (cocaine cues vs. neutral cocaine cues and negative emotional cues vs. neutral emotional cues) on activity in the dorsal striatum, ventral striatum (VS), amygdala, and dorsal anterior cingulate cortex (dACC). RESULTS: There were no significant sex or group differences in cocaine cue-reactivity in any of the ROIs. Results did reveal significant emotional cue-reactivity in the amygdala and VS, but these effects were not moderated by group or sex. Exploratory analyses demonstrated that emotional cue-induced activation of the dACC and VS was negatively associated with years of regular cocaine use in female CUs, while this relationship was absent in male CUs. CONCLUSIONS: While speculative, the sex-specific associations between years of regular use and emotional cue-reactivity in the dACC and VS suggest that, with longer years of use, female CUs become less sensitive to aversive stimuli, including the negative consequences of cocaine use, which could account for the observed "telescoping effect" in female CUs.


Subject(s)
Cocaine-Related Disorders , Cues , Emotions , Humans , Male , Female , Cocaine-Related Disorders/psychology , Cocaine-Related Disorders/physiopathology , Adult , Emotions/physiology , Magnetic Resonance Imaging , Sex Characteristics , Cocaine/pharmacology , Young Adult , Amygdala/diagnostic imaging , Amygdala/physiopathology , Gyrus Cinguli/physiopathology , Gyrus Cinguli/diagnostic imaging , Brain/diagnostic imaging , Sex Factors , Case-Control Studies
16.
Pharmacol Biochem Behav ; 240: 173771, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38670466

ABSTRACT

In the United States, the societal costs associated with drug use surpass $500 billion annually. The rewarding and reinforcing properties that drive the use of these addictive substances are typically examined concerning the neurobiological effects responsible for their abuse potential. In this review, terms such as "abuse potential," "drug," and "addictive properties" are used due to their relevance to the methodological, theoretical, and conceptual framework for understanding the phenomenon of drug-taking behavior and the associated body of preclinical and clinical literature. The use of these terms is not intended to cast aspersions on individuals with substance use disorders (SUD). Understanding what motivates substance use has been a focus of SUD research for decades. Much of this corpus of work has focused on the shared effects of each drug class to increase dopaminergic transmission within the central reward pathways of the brain, or the "reward center." However, the precise influence of each drug class on dopamine signaling, and the extent thereof, differs considerably. Furthermore, the aforementioned substances have effects on several neurobiological targets that mediate and modulate their addictive properties. The current manuscript sought to review the influence of drug class on the rewarding effects of each of the major pharmacological classes of addictive drugs (i.e., psychostimulants, opioids, nicotine, alcohol, and cannabinoids). Our review suggests that even subtle differences in drug effects can result in significant variability in the subjective experience of the drug, altering rewarding and other reinforcing effects. Additionally, this review will argue that reward (i.e., the attractive and motivational property of a stimulus) alone is not sufficient to explain the abuse liability of these substances. Instead, abuse potential is best examined as a function of both positive and negative reinforcing drug effects (i.e., stimuli that the subject will work to attain and stimuli that the subject will work to end or avoid, respectively). Though reward is central to drug use, the factors that motivate and maintain drug taking are varied and complex, with much to be elucidated.


Subject(s)
Reward , Substance-Related Disorders , Humans , Substance-Related Disorders/psychology , Animals , Behavior, Addictive/psychology , Dopamine/metabolism , Brain/drug effects , Brain/metabolism , Cannabinoids/pharmacology , Motivation , Nicotine/pharmacology , Reinforcement, Psychology , Analgesics, Opioid/pharmacology , Cocaine/pharmacology
17.
Article in English | MEDLINE | ID: mdl-38649130

ABSTRACT

Cocaine use disorder (CUD) is a global health problem with no approved medications. One potential treatment target is the gut microbiome, but it is unknown if cocaine induces long-lasting effects on gut microbes. A novel therapeutic candidate for CUD, cannabidiol (CBD), can improve gut function in rodent models. It is possible that protective effects of CBD against cocaine use are mediated by improving gut health. We examined this question in this experiment. Cocaine conditioned place preference (CPP) was conducted in adult male C57BL/6JArc mice. Mice were treated with vehicle or 20 mg/kg CBD prior to all cocaine CPP sessions (N = 11-13/group). Mice were tested drug free 1, 14 and 28 days after cessation of cocaine and CBD treatment. Fecal samples were collected prior to drug treatment and after each test session. Gut microbiome analyses were conducted using 16 s rRNA sequencing and correlated with behavioural parameters. We found a persistent preference for a cocaine-environment in mice, and long-lasting changes to gut microbe alpha diversity. Cocaine caused persistent changes to beta diversity which lasted for 4 weeks. CBD treatment reduced cocaine-environment preference during abstinence from cocaine and returned gut beta diversity measures to control levels. CBD treatment increased the relative abundance of Firmicutes phyla and Oscillospira genus, but decreased Bacteroidetes phyla and Bacteroides acidifaciens species. Preference score in cocaine-treated mice was positively correlated with abundance of Actinobacteria, whereas in mice treated with CBD and cocaine, the preference score was negatively correlated with Tenericutes abundance. Here we show that CBD facilitates cocaine extinction memory and reverses persistent cocaine-induced changes to gut microbe diversity. Furthermore, CBD increases the abundance of gut microbes which have anti-inflammatory properties. This suggests that CBD may act via the gut to reduce the memory of cocaine reward. Our data suggest that improving gut health and using CBD could limit cocaine abuse.


Subject(s)
Cannabidiol , Cocaine , Extinction, Psychological , Gastrointestinal Microbiome , Mice, Inbred C57BL , Animals , Cannabidiol/pharmacology , Male , Gastrointestinal Microbiome/drug effects , Cocaine/pharmacology , Mice , Extinction, Psychological/drug effects , Cocaine-Related Disorders/drug therapy
18.
Behav Neurosci ; 138(2): 108-124, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38661670

ABSTRACT

The cannabinoid system is being researched as a potential pharmaceutical target for a multitude of disorders. The present study examined the effect of indirect and direct cannabinoid agonists on mesolimbic dopamine release and related behaviors in C57BL/6J (B6) mice. The indirect cannabinoid agonist N-arachidonoyl serotonin (AA-5-HT) indirectly agonizes the cannabinoid system by preventing the metabolism of endocannabinoids through fatty acid amide hydrolase inhibition while also inhibiting transient receptor potential vanilloid Type 1 channels. Effects of AA-5-HT were compared with the direct cannabinoid receptor Type 1 agonist arachidonoyl-2'-chloroethylamide (ACEA). In Experiment 1, mice were pretreated with seven daily injections of AA-5-HT, ACEA, or vehicle prior to assessments of locomotor activity using open field (OF) testing and phasic dopamine release using in vivo fixed potential amperometry. Chronic exposure to AA-5-HT did not alter locomotor activity or mesolimbic dopamine functioning. Chronic exposure to ACEA decreased rearing and decreased phasic dopamine release while increasing the dopaminergic response to cocaine. In Experiment 2, mice underwent AA-5-HT, ACEA, or vehicle conditioned place preference, then saccharin preference testing, a measure commonly associated with anhedonia. Mice did not develop a conditioned place preference or aversion for AA-5-HT or ACEA, and repeated exposure to AA-5-HT or ACEA did not alter saccharin preference. Altogether, the findings suggest that neither of these drugs induce behaviors that are classically associated with abuse liability in mice; however, direct cannabinoid receptor Type 1 agonism may play more of a role in mediating mesolimbic dopamine functioning than indirect cannabinoid agonism. (PsycInfo Database Record (c) 2024 APA, all rights reserved).


Subject(s)
Cannabinoid Receptor Agonists , Dopamine , Mice, Inbred C57BL , Animals , Dopamine/metabolism , Male , Mice , Cannabinoid Receptor Agonists/pharmacology , Serotonin/metabolism , Locomotion/drug effects , Behavior, Animal/drug effects , Arachidonic Acids/pharmacology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Cocaine/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/metabolism , Motor Activity/drug effects
19.
J Neurosci ; 44(23)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38637154

ABSTRACT

Cocaine use disorder is a significant public health issue without an effective pharmacological treatment. Successful treatments are hindered in part by an incomplete understanding of the molecular mechanisms that underlie long-lasting maladaptive plasticity and addiction-like behaviors. Here, we leverage a large RNA sequencing dataset to generate gene coexpression networks across six interconnected regions of the brain's reward circuitry from mice that underwent saline or cocaine self-administration. We identify phosphodiesterase 1b (Pde1b), a Ca2+/calmodulin-dependent enzyme that increases cAMP and cGMP hydrolysis, as a central hub gene within a nucleus accumbens (NAc) gene module that was bioinformatically associated with addiction-like behavior. Chronic cocaine exposure increases Pde1b expression in NAc D2 medium spiny neurons (MSNs) in male but not female mice. Viral-mediated Pde1b overexpression in NAc reduces cocaine self-administration in female rats but increases seeking in both sexes. In female mice, overexpressing Pde1b in D1 MSNs attenuates the locomotor response to cocaine, with the opposite effect in D2 MSNs. Overexpressing Pde1b in D1/D2 MSNs had no effect on the locomotor response to cocaine in male mice. At the electrophysiological level, Pde1b overexpression reduces sEPSC frequency in D1 MSNs and regulates the excitability of NAc MSNs. Lastly, Pde1b overexpression significantly reduced the number of differentially expressed genes (DEGs) in NAc following chronic cocaine, with discordant effects on gene transcription between sexes. Together, we identify novel gene modules across the brain's reward circuitry associated with addiction-like behavior and explore the role of Pde1b in regulating the molecular, cellular, and behavioral responses to cocaine.


Subject(s)
Cocaine-Related Disorders , Cyclic Nucleotide Phosphodiesterases, Type 1 , Gene Regulatory Networks , Mice, Inbred C57BL , Nucleus Accumbens , Sex Characteristics , Animals , Male , Female , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Mice , Cocaine-Related Disorders/genetics , Cocaine-Related Disorders/metabolism , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/genetics , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats , Cocaine/pharmacology , Reward
20.
Sci Signal ; 17(832): eadl4738, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38626009

ABSTRACT

Cocaine use disorder (CUD) is a chronic neuropsychiatric condition that results from enduring cellular and molecular adaptations. Among substance use disorders, CUD is notable for its rising prevalence and the lack of approved pharmacotherapies. The nucleus accumbens (NAc), a region that is integral to the brain's reward circuitry, plays a crucial role in the initiation and continuation of maladaptive behaviors that are intrinsic to CUD. Leveraging advancements in neuroproteomics, we undertook a proteomic analysis that spanned membrane, cytosolic, nuclear, and chromatin compartments of the NAc in a mouse model. The results unveiled immediate and sustained proteomic modifications after cocaine exposure and during prolonged withdrawal. We identified congruent protein regulatory patterns during initial cocaine exposure and reexposure after withdrawal, which contrasted with distinct patterns during withdrawal. Pronounced proteomic shifts within the membrane compartment indicated adaptive and long-lasting molecular responses prompted by cocaine withdrawal. In addition, we identified potential protein translocation events between soluble-nuclear and chromatin-bound compartments, thus providing insight into intracellular protein dynamics after cocaine exposure. Together, our findings illuminate the intricate proteomic landscape that is altered in the NAc by cocaine use and provide a dataset for future research toward potential therapeutics.


Subject(s)
Cocaine-Related Disorders , Cocaine , Mice , Animals , Nucleus Accumbens/metabolism , Proteomics , Cocaine/pharmacology , Cocaine-Related Disorders/genetics , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/psychology , Chromatin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL