Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 215
Filter
1.
Proc Natl Acad Sci U S A ; 119(25): e2122477119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35700362

ABSTRACT

Alcohol intoxication at early ages is a risk factor for the development of addictive behavior. To uncover neuronal molecular correlates of acute ethanol intoxication, we used stable-isotope-labeled mice combined with quantitative mass spectrometry to screen more than 2,000 hippocampal proteins, of which 72 changed synaptic abundance up to twofold after ethanol exposure. Among those were mitochondrial proteins and proteins important for neuronal morphology, including MAP6 and ankyrin-G. Based on these candidate proteins, we found acute and lasting molecular, cellular, and behavioral changes following a single intoxication in alcohol-naïve mice. Immunofluorescence analysis revealed a shortening of axon initial segments. Longitudinal two-photon in vivo imaging showed increased synaptic dynamics and mitochondrial trafficking in axons. Knockdown of mitochondrial trafficking in dopaminergic neurons abolished conditioned alcohol preference in Drosophila flies. This study introduces mitochondrial trafficking as a process implicated in reward learning and highlights the potential of high-resolution proteomics to identify cellular mechanisms relevant for addictive behavior.


Subject(s)
Alcoholic Intoxication , Dopaminergic Neurons , Ethanol , Hippocampus , Nerve Tissue Proteins , Alcoholic Intoxication/metabolism , Alcoholic Intoxication/pathology , Animals , Behavior, Addictive/chemically induced , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dose-Response Relationship, Drug , Drosophila melanogaster , Ethanol/administration & dosage , Ethanol/toxicity , Gene Knockdown Techniques , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mitochondria/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Transport/drug effects
2.
J Ethnopharmacol ; 284: 114824, 2022 Feb 10.
Article in English | MEDLINE | ID: mdl-34763040

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Mitragyna speciosa (Korth.) Havil., popularly known as Kratom (KT), is a medicinal plant used for pain suppression in Southeast Asia. It has been claimed to assist drug users withdraw from methamphetamine (METH) dependence. However, its use was controversial and not approved yet. AIM OF THE STUDY: This study was conducted to characterize local field potential (LFP) patterns in the nucleus accumbens (NAc) and the hippocampus (HP) in mice with METH conditioned place preference (CPP) that were treated with KT alkaloid extract. MATERIALS AND METHODS: Male Swiss albino ICR mice were implanted with intracraneal electrodes into the NAc and HP. To induce METH CPP, animals were injected intraperitoneally once a day with METH (1 mg/kg) and saline (0.9% w/v) alternately and put into METH/saline compartments to experience the associations between drug/saline injection and the unique environmental contexts for 10 sessions. Control group received saline injection paired with both saline/saline compartments. On post-conditioning day, effects of 40 (KT40), 80 (KT80) mg/kg KT alkaloid extract and 20 mg/kg bupropion (BP) on CPP scores and LFP powers and NAc-HP coherence were tested. RESULTS: Two-way ANOVA revealed significant induction of CPP by METH sessions (P < 0.01). Multiple comparisons indicated that METH CPP was completely abolished by KT80 (P < 0.001). NAc gamma I (30.0-44.9 Hz) and HP delta (1.0-3.9 Hz) powers were significantly increased in mice with METH CPP (P < 0.01). The elevated NAc gamma I was significantly suppressed by KT80 (P < 0.05) and the increased HP delta was significantly reversed by KT40 (P < 0.01) and KT80 (P < 0.001). In addition, NAc-HP coherence was also significantly increased in gamma I (30.0-44.9 Hz) frequency range (P < 0.05) but it was reversed by KT80 (P < 0.05). Treatment with BP did not produce significant effect on these parameters. CONCLUSIONS: These findings demonstrated that KT alkaloid extract significantly reversed CPP scores and LFP patterns induced by METH administration. The ameliorative effects of the extract might be beneficial for treatment of METH craving and addiction.


Subject(s)
Alkaloids/pharmacology , Behavior, Addictive/drug therapy , Methamphetamine/adverse effects , Mitragyna/chemistry , Plant Extracts/pharmacology , Plant Leaves/chemistry , Alkaloids/chemistry , Animals , Behavior, Addictive/chemically induced , Central Nervous System Stimulants/adverse effects , Humans , Male , Mice , Mice, Inbred ICR , Nucleus Accumbens/drug effects , Phytotherapy , Plant Extracts/chemistry
3.
PLoS One ; 16(6): e0252516, 2021.
Article in English | MEDLINE | ID: mdl-34061895

ABSTRACT

Gambling Disorder (GD) has recently been reclassified from an impulse-control disorder to a behavioural addiction and, as in other addictive disorders, the dopaminergic reward system is involved. According to neuroimaging studies, alterations within the striatal dopaminergic signalling can occur in GD. However, the findings to date are controversial and there has been no agreement yet on how the reward system is affected on a molecular basis. Within the last 20 years, there has been growing evidence for a higher risk to develop GD in response to certain dopaminergic medication. Especially the dopamine agonists pramipexole and ropinirole, and the dopamine modulator aripiprazole seem to increase the likelihood for GD. The goal of this study was to examine the association between a prescription for either of the three pharmaceuticals and a GD diagnosis in a large cross-sectional study of the Swedish population. Compared to patients with any other dopaminergic drug prescription (38.7% with GD), the diagnosis was more common in patients with a dopamine agonist prescription (69.8% with GD), resulting in an odds ratio of 3.2. A similar association was found between aripiprazole prescriptions and GD diagnoses, which were analysed within the subgroup of all patients with schizophrenia or a schizotypal, delusional, or another non-mood psychotic disorder. An aripiprazole prescription increased the likelihood of GD (88.8%) in comparison to patients without an aripiprazole prescription (71.2%) with an odds ratio of 3.4. This study contributes to the increasingly reliable evidence for an association between several dopaminergic drugs and a higher risk for developing GD. Therefore, one future research goal should be a better understanding of the neurobiology in GD to be able to design more selective dopaminergic medication with less severe side effects. Additionally, this knowledge could enable the development of pharmacotherapy in GD and other addictive disorders.


Subject(s)
Aripiprazole/adverse effects , Behavior, Addictive/chemically induced , Dopamine Agonists/adverse effects , Gambling/chemically induced , Indoles/adverse effects , Pramipexole/adverse effects , Registries , Adult , Aged , Aged, 80 and over , Behavior, Addictive/diagnosis , Behavior, Addictive/epidemiology , Behavior, Addictive/metabolism , Cross-Sectional Studies , Dopamine/metabolism , Female , Gambling/diagnosis , Gambling/epidemiology , Gambling/metabolism , Humans , Male , Middle Aged , Reward , Risk Factors , Sweden/epidemiology , Young Adult
4.
Br J Pharmacol ; 178(19): 3869-3887, 2021 10.
Article in English | MEDLINE | ID: mdl-33987827

ABSTRACT

BACKGROUND AND PURPOSE: Methoxphenidine is a dissociative-based novel psychoactive designer drug. Although fatal accidents from methoxphenidine abuse have been reported, recreational use of the drug continues. We aim to provide scientific supportfor legal regulation of recreational abuse of methoxphenidine by demonstrating its the pharmacological action. EXPERIMENTAL APPROACH: Addictive potential of methoxphenidine was examined using intravenous self-administration test with rats and conditioned place preference test with mice. Further, a series of behavioural tests (open field test, elevated plus maze test, novel object recognition test, social interaction test and tail suspension test) performed to assess whether methoxphenidine caused schizophrenia-related symptoms in mice. Additionally, neurotransmitter enzyme-linked immunosorbent assay and western blot were used to confirm methoxphenidine-induced neurochemical changes in specific brain regions related to abnormal behaviours. KEY RESULTS: Methoxphenidine caused addictive behaviours via reinforcing and rewarding effects. Consistently, methoxphenidine induced over-activation of dopamine pathways in the nuclear accumbens, indicating activation of the brain reward circuit. Also, methoxphenidine caused all categories of schizophrenia-related symptoms, including positive symptoms (hyperactivity, impulsivity), negative symptoms (anxiety, social withdrawal, depression) and cognitive impairment. Consistently, methoxphenidine led to the disruption of the hippocampal-prefrontal cortex pathway that is considered to be pathological involved in schizophrenia. CONCLUSIONS AND IMPLICATIONS: We demonastrate that methoxphenidine causes addictive and schizophrenia-like behaviours and induces neurochemical changes in brain regions associated with these behaviours. We propose that methoxphenidine could be used in developing useful animal disease models and that it also requires legal restrictions on its recreational use.


Subject(s)
Behavior, Addictive , Schizophrenia , Animals , Behavior, Addictive/chemically induced , Brain , Mice , Piperidines , Rats
6.
Mol Psychiatry ; 26(1): 234-246, 2021 01.
Article in English | MEDLINE | ID: mdl-32071384

ABSTRACT

The development of drug addiction is associated with functional adaptations within the reward circuitry, within which the nucleus accumbens (NAc) is anatomically positioned as an interface between motivational salience and behavioral output. The functional output of NAc is profoundly altered after exposure to drugs of abuse, and some of the functional changes continue to evolve during drug abstinence, contributing to numerous emotional and motivational alterations related drug taking, seeking, and relapse. As in most brain regions, the functional output of NAc is critically dependent on the dynamic interaction between excitation and inhibition. One of the most prominent sources of inhibition within the NAc arises from fast-spiking interneurons (FSIs). Each NAc FSI innervates hundreds of principal neurons, and orchestrates population activity through its powerful and sustained feedforward inhibition. While the role of NAc FSIs in the context of drug addiction remains poorly understood, emerging evidence suggests that FSIs and FSI-mediated local circuits are key targets for drugs of abuse to tilt the functional output of NAc toward a motivational state favoring drug seeking and relapse. In this review, we discuss recent findings and our conceptualization about NAc FSI-mediated regulation of motivated and cocaine-induced behaviors. We hope that the conceptual framework proposed in this review may provide a useful guidance for ongoing and future studies to determine how FSIs influence the function of NAc and related reward circuits, ultimately leading to addictive behaviors.


Subject(s)
Behavior, Addictive/metabolism , Interneurons/metabolism , Motivation , Nucleus Accumbens/metabolism , Behavior, Addictive/chemically induced , Cocaine/pharmacology , Cocaine-Related Disorders/metabolism , Drug-Seeking Behavior/drug effects , Humans
7.
Psychopharmacology (Berl) ; 238(1): 305-319, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33111197

ABSTRACT

RATIONALE: Initial exposure to cannabinoids, including Δ-9-tetrahydrocannabinol (THC), often occurs during adolescence. Considerable neurodevelopmental alterations occur throughout adolescence, and the environmental insult posed by exogenous cannabinoid exposure may alter natural developmental trajectories. Multiple studies suggest that long-lasting deficits in cognitive function occur as a result of adolescent cannabis use, but considerable variability exists in the magnitude of these effects. OBJECTIVES: We sought to establish a novel procedure for achieving intravenous THC self-administration in adolescent rats in order to determine if volitional THC intake in adolescence produced indices of addiction-related behavior, altered working memory performance in adulthood, or altered the expression of proteins associated with these behaviors across several brain regions. METHODS: Male and female adolescent rats learned to operantly self-administer escalating doses of THC intravenously from PD 32-51. Upon reaching adulthood they were tested in abstinence for cued reinstatement of THC-seeking and working memory performance on a delayed-match-to-sample task. In a separate cohort, glutamatergic, GABAergic, and cannabinoid receptor protein expression was measured in multiple brain regions. RESULTS: Both male and female adolescents self-administered THC and exhibited cue-induced lever pressing throughout abstinence. THC-exposed males exhibited slightly enhanced working memory performance in adulthood, and better performance positively correlated with total THC self-administered during adolescence. Adolescent THC-exposed rats exhibited reductions in CB1, GABA, and glutamate receptor protein, primarily in the prefrontal cortex, dorsal hippocampus, and ventral tegmental area. CONCLUSIONS: These results suggest that THC exposure at self-administered doses can produce moderate behavioral and molecular alterations, including sex-dependent effects on working memory performance in adulthood.


Subject(s)
Aging/drug effects , Behavior, Addictive/chemically induced , Brain/drug effects , Dronabinol/toxicity , Memory, Short-Term/drug effects , Receptors, Cell Surface/metabolism , Aging/metabolism , Aging/psychology , Animals , Behavior, Addictive/metabolism , Behavior, Addictive/psychology , Brain/metabolism , Dronabinol/administration & dosage , Female , Injections, Intravenous , Male , Marijuana Smoking/adverse effects , Marijuana Smoking/psychology , Rats , Receptors, Cell Surface/genetics , Self Administration , Sex Characteristics
8.
Ann N Y Acad Sci ; 1489(1): 3-16, 2021 04.
Article in English | MEDLINE | ID: mdl-32147860

ABSTRACT

The treatment of substance use disorders has always been challenging because multiple neurotransmitters mediate addiction. However, with smoking being the leading cause of preventable death and the recent opioid epidemic in the United States, the search for novel solutions becomes more imperative. In this review, we discuss the use of antibodies to treat addictions and highlight areas of success and areas that require improvement, using examples from cocaine, nicotine, and opioid vaccines. Through each example, we examine creative problem-solving strategies for developing future vaccines, such as using an adenovirus vector as a carrier, designing bivalent vaccines, stimulating Toll-like receptors for adjuvant effects, and altering the route of administration. Our review also covers passive immunization alone to override or prevent drug toxicity as well as in combination with vaccines for more rapid and potentially greater efficacy.


Subject(s)
Behavior, Addictive/therapy , Immunotherapy , Substance-Related Disorders/therapy , Analgesics, Opioid/toxicity , Behavior, Addictive/chemically induced , Behavior, Addictive/immunology , Cocaine/toxicity , Humans , Immunization, Passive/methods , Substance-Related Disorders/immunology , Substance-Related Disorders/prevention & control , United States/epidemiology
9.
Drugs ; 80(15): 1509-1524, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32776315

ABSTRACT

Opioid use disorder (OUD) represents a major public health problem that affects millions of people in the USA and worldwide. The relapsing and recurring aspect of OUD, driven by lasting neurobiological adaptations at different reward centres in the brain, represents a major obstacle towards successful long-term remission from opioid use. Currently, three drugs that modulate the function of the opioidergic receptors, methadone, buprenorphine and naltrexone have been approved by the US Food and Drug Administration (FDA) to treat OUD. In this review, we discuss the limitations and challenges associated with the current maintenance and medication-assisted withdrawal strategies commonly used to treat OUD. We further explore the involvement of glutamatergic, endocannabinoid and orexin signaling systems in the development, maintenance and expression of addiction-like behaviours in animal models of opioid addiction, and as potential and novel targets to expand therapeutic options to treat OUD. Despite a growing preclinical literature highlighting the role of these potential targets in animal models of opioid addiction, clinical and translational studies for novel treatments of OUD remain limited and inconclusive. Further preclinical and clinical investigations are needed to expand the arsenal of primary treatment options and adjuncts to maximise efficacy and prevent relapse.


Subject(s)
Analgesics, Opioid/adverse effects , Behavior, Addictive/drug therapy , Narcotic Antagonists/therapeutic use , Opiate Substitution Treatment/methods , Opioid-Related Disorders/drug therapy , Animals , Behavior, Addictive/chemically induced , Behavior, Addictive/physiopathology , Behavior, Addictive/psychology , Brain/drug effects , Brain/physiopathology , Buprenorphine/pharmacology , Buprenorphine/therapeutic use , Disease Models, Animal , Endocannabinoids/metabolism , Glutamic Acid/metabolism , Humans , Methadone/pharmacology , Methadone/therapeutic use , Naltrexone/pharmacology , Naltrexone/therapeutic use , Narcotic Antagonists/pharmacology , Opioid-Related Disorders/etiology , Opioid-Related Disorders/physiopathology , Opioid-Related Disorders/psychology , Orexins/metabolism , Reward , Secondary Prevention/methods , Signal Transduction/drug effects , Signal Transduction/physiology , Treatment Outcome
10.
Neuropharmacology ; 176: 108243, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32702403

ABSTRACT

Most people start experimenting with tobacco products or e-cigarettes in early adolescence and become habitual smokers in late adolescence or adulthood. These studies investigated if exposure to tobacco smoke or nicotine during early and mid-adolescence affects nicotine intake in late adolescence and early adulthood. Male and female rats were exposed to tobacco smoke from low- and high-nicotine SPECTRUM cigarettes or nicotine (0.3 mg/kg, twice a day) from postnatal day (P) 24-42. The self-administration sessions started at P55. The rats self-administered nicotine for 14-15 days under a fixed-ratio 1 schedule, and on the first day, the maximum number of infusions was twenty. Exposure to smoke from high, but not low, nicotine cigarettes during adolescence increased nicotine self-administration in female but not male rats. Adolescent treatment with nicotine facilitated nicotine self-administration. On the first day of nicotine self-administration, nicotine-treated rats reached the maximum number of infusions before the saline-treated control rats. Nicotine intake was also higher in the nicotine-treated female rats than in the saline-treated females. There was no sex difference in nicotine intake in controls when the data from the studies were combined. Smoke exposure led to a dose-dependent increase in plasma nicotine and cotinine levels in adolescent rats. Exposure to smoke from high-nicotine cigarettes and 0.3 mg/kg of nicotine led to plasma nicotine and cotinine levels that are similar to those in tobacco users. These findings indicate that in females, but not males, exposure to nicotine during adolescence may facilitate smoking and e-cigarette use later in life.


Subject(s)
Behavior, Addictive/chemically induced , Inhalation Exposure/adverse effects , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Tobacco Smoke Pollution/adverse effects , Age Factors , Animals , Behavior, Addictive/blood , Behavior, Addictive/psychology , Female , Male , Nicotine/blood , Nicotinic Agonists/blood , Rats , Self Administration
11.
Behav Brain Res ; 390: 112624, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32428634

ABSTRACT

It has been reported that environmental stimuli can positively influence addictive responses and the pharmacological effects of drugs of abuse. In this work, we evaluated the ability of environmental enrichment (EE) to attenuate addictive behaviours in mice after repeated exposure to toluene. We also analysed the role of D1 receptors (D1R) in animals chronically exposed to toluene and in those housed under EE. Mice (Swiss Webster) were exposed to toluene (0, 2000 or 4000 ppm, 30 min a day), and addictive responses were examined in the behavioural sensitization model. The induction of sensitization was evaluated over 4 weeks, and its expression was assessed in animals repeatedly exposed to toluene (0 or 4000 ppm, 30 min a day/4 weeks) and then housed under EE conditions during 4 more weeks. D1R levels were measured under these two experimental conditions in the prefrontal cortex, nucleus accumbens, hippocampus and caudate. The results showed that D1R levels decreased during toluene-induced behavioural sensitization. An increase in D1R levels was found in animals housed in EE conditions, in addition to the attenuated expression of behavioural sensitization. These results indicate that environmental stimulation attenuates addictive behaviour induced by toluene and that dynamic changes in D1R are linked in this response.


Subject(s)
Behavior, Addictive/chemically induced , Behavior, Addictive/prevention & control , Behavior, Animal/drug effects , Central Nervous System Sensitization/drug effects , Cerebrum/metabolism , Environment , Receptors, Dopamine D1/metabolism , Toluene/pharmacology , Animals , Caudate Nucleus/metabolism , Disease Models, Animal , Hippocampus/metabolism , Male , Mice , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Toluene/administration & dosage
12.
Proc Natl Acad Sci U S A ; 117(18): 9991-10002, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32312805

ABSTRACT

The initial response to an addictive substance can facilitate repeated use: That is, individuals experiencing more positive effects are more likely to use that drug again. Increasing evidence suggests that psychoactive cannabinoid use in adolescence enhances the behavioral effects of cocaine. However, despite the behavioral data, there is no neurobiological evidence demonstrating that cannabinoids can also alter the brain's initial molecular and epigenetic response to cocaine. Here, we utilized a multiomics approach (epigenomics, transcriptomics, proteomics, and phosphoproteomics) to characterize how the rat brain responds to its first encounter with cocaine, with or without preexposure to the synthetic cannabinoid WIN 55,212-2 (WIN). We find that in adolescent (but not in adult) rats, preexposure to WIN results in cross-sensitization to cocaine, which correlates with histone hyperacetylation and decreased levels of HDAC6 in the prefrontal cortex (PFC). In the PFC, we also find that WIN preexposure blunts the typical mRNA response to cocaine and instead results in alternative splicing and chromatin accessibility events, involving genes such as Npas2 Moreover, preexposure to WIN enhances the effects of cocaine on protein phosphorylation, including ERK/MAPK-targets like gephyrin, and modulates the synaptic AMPAR/GluR composition both in the PFC and the nucleus accumbens (NAcc). PFC-NAcc gene network topological analyses, following cocaine exposure, reveal distinct top nodes in the WIN preexposed group, which include PACAP/ADCYAP1. These preclinical data demonstrate that adolescent cannabinoid exposure reprograms the initial behavioral, molecular, and epigenetic response to cocaine.


Subject(s)
Behavior, Addictive/genetics , Behavior, Animal/drug effects , Cannabinoids/adverse effects , Cocaine/adverse effects , Adolescent , Animals , Behavior, Addictive/chemically induced , Behavior, Addictive/pathology , Benzoxazines/adverse effects , Benzoxazines/pharmacology , Cannabinoids/pharmacology , Circadian Rhythm Signaling Peptides and Proteins/genetics , Cocaine/pharmacology , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Gene Expression Regulation/drug effects , Histone Deacetylase 6/genetics , Humans , Membrane Proteins/pharmacology , Morpholines/adverse effects , Morpholines/pharmacology , Naphthalenes/adverse effects , Naphthalenes/pharmacology , Phosphoproteins/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Prefrontal Cortex/drug effects , Proteome/drug effects , Rats , Transcriptome/drug effects
13.
Arch Toxicol ; 94(7): 2505-2516, 2020 07.
Article in English | MEDLINE | ID: mdl-32296860

ABSTRACT

The use of new psychoactive substances (NPSs) as a substitute for illegal drugs is increasing rapidly and is a serious threat to public health. 25C-NBF is a newly synthesized phenethylamine-type NPS that acts as a 5-hydroxyindoleacetic acid (5-HT) receptor agonist, but little is known about its pharmacological effects. Considering that NPSs have caused unexpected harmful effects leading to emergency and even death, scientific confirmation of the potential adverse effects of 25C-NBF is essential. In the present study, we investigated whether 25C-NBF has addictive and neurotoxic potential and causes neurochemical changes. In addictive potential assessments, high conditioned place preference (CPP) scores and stable self-administration (SA) were observed in the 25C-NBF groups (CPP [3 mg kg-1]; SA [0.01, 0.03, 0.1 mg kg-1]), suggesting the addictive liability of 25C-NBF. In neurotoxic potential assessments, 25C-NBF treatment (single super-high dose [1 × 15, 30, 40 mg kg-1]; repeated high dose [4 × 8, 15, 30 mg kg-1]) resulted in reduced motor activity (open field test), abnormal motor coordination (rota-rod test) and impaired recognition memory (novel object recognition test), suggesting that 25C-NBF is neurotoxic leading to motor impairment and memory deficits. Subsequently, immunohistochemistry showed that 25C-NBF treatment decreased tyrosine hydroxylase (TH) expression and increased ionized calcium-binding adapter molecule 1 (Iba-1) expression in the striatum. Taken together, our results clearly demonstrate the dangers of recreational use of 25C-NBF, and we suggest that people stop using 25C-NBF and other NPSs whose pharmacological effects are not precisely known.


Subject(s)
Behavior, Addictive/chemically induced , Behavior, Animal/drug effects , Brain/drug effects , Neurotoxicity Syndromes/etiology , Phenethylamines/toxicity , Psychotropic Drugs/toxicity , Substance-Related Disorders/etiology , Animals , Behavior, Addictive/metabolism , Behavior, Addictive/psychology , Brain/metabolism , Brain/physiopathology , Calcium-Binding Proteins/metabolism , Conditioning, Psychological/drug effects , Glial Fibrillary Acidic Protein/metabolism , Locomotion/drug effects , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Microfilament Proteins/metabolism , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/physiopathology , Open Field Test/drug effects , Rats, Sprague-Dawley , Rotarod Performance Test , Substance-Related Disorders/metabolism , Substance-Related Disorders/psychology , Tyrosine 3-Monooxygenase/metabolism
14.
Neuropharmacology ; 168: 108007, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32092436

ABSTRACT

The orbitofrontal cortex (OFC) plays a fundamental role in motivated behavior and decision-making. In humans, OFC structure and function is significantly disrupted in drug using and dependent individuals, including those exhibiting chronic alcohol use and alcoholism. In animal models, the OFC has been shown to significantly influence the seeking of non-alcohol drugs of abuse. However direct investigations of the OFC during alcohol seeking and use have been more limited. In the studies reported here, we inactivated lateral (lOFC) or medial OFC (mOFC) subregions in rats during multiple stages of alcohol seeking. After one month of intermittent access to homecage 20% ethanol (EtOH), rats were trained to self-administer EtOH under an FR3 schedule and implanted with cannulae directed to lOFC or mOFC. We inactivated OFC subregions with baclofen/muscimol during EtOH self-administration, extinction, cue-induced reinstatement, and progressive ratio testing to broadly characterize the influence of these subregions on alcohol seeking. There were no significant effects of mOFC or lOFC inactivation during FR3 self-administration, extinction, or progressive ratio self-administration. However, lOFC, and not mOFC, inactivation significantly decreased cue-induced reinstatement of EtOH seeking. These findings contribute new information to the specific impact of OFC manipulation on operant alcohol seeking, support previous studies investigating the role of OFC in seeking and consumption of alcohol and other drugs of abuse, and indicate a specific role for lOFC vs. mOFC in reinstatement.


Subject(s)
Alcohol Drinking/metabolism , Behavior, Addictive/metabolism , Ethanol/administration & dosage , GABA Agonists/administration & dosage , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Alcohol Drinking/psychology , Animals , Behavior, Addictive/chemically induced , Behavior, Addictive/psychology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Injections, Intravenous , Male , Rats , Rats, Long-Evans , Self Administration
15.
Pharmacol Biochem Behav ; 191: 172880, 2020 04.
Article in English | MEDLINE | ID: mdl-32105663

ABSTRACT

Caffeine is the most widely consumed psychoactive substance in the world. However, there is controversy about whether becoming addicted to caffeine is possible and a lack of well-established animal models to examine caffeine consumption. The present study sought to establish a model of caffeine consumption in Wistar rats, identify different rat populations based on caffeine preference, and determine whether extended voluntary caffeine consumption produces compulsive-like caffeine intake and withdrawal symptoms. Male Wistar rats were used throughout the experiment. The optimal concentration of caffeine to maximize caffeine consumption and caffeine preference was determined. Rats were then given continuous access to caffeine, followed by intermittent access. Rats were tested for signs of withdrawal-like behavior by measuring mechanical nociception and irritability-like behavior. Rats were further examined for compulsive-like caffeine consumption using quinine adulteration. Dose-response testing indicated an optimal caffeine concentration of 0.3 mg/mL. During intermittent access to caffeine, the rats did not escalate their caffeine intake and instead exhibited a decrease in intake over sessions. Three groups of rats were identified based on caffeine preference (high, medium, and low) across continuous and intermittent access. These three groups of rats matched low (1 cup), medium (2 cups), and high (4 cups) levels of daily coffee consumption in humans. Caffeine-consuming rats did not exhibit differences in mechanical nociception or irritability-like behavior compared with controls. In high caffeine-preferring rats but not in medium or low caffeine-preferring rats, compulsive-like caffeine consumption was observed. The present study established a rodent model of caffeine consumption that resulted in large individual differences in caffeine intake, similar to humans. Compulsive-like caffeine consumption in high caffeine-preferring rats and differences in caffeine preference between groups suggest that caffeine may result in compulsive-like intake in a subpopulation of subjects. Further testing is necessary to determine the factors that contribute to differences in caffeine preference and compulsive-like intake.


Subject(s)
Behavior, Addictive/chemically induced , Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Individuality , Animals , Behavior, Animal/drug effects , Caffeine/administration & dosage , Choice Behavior/drug effects , Dose-Response Relationship, Drug , Irritable Mood/drug effects , Male , Nociception/drug effects , Quinine/pharmacology , Rats , Rats, Wistar , Substance Withdrawal Syndrome
16.
Neuropharmacology ; 166: 107974, 2020 04.
Article in English | MEDLINE | ID: mdl-32007624

ABSTRACT

On the basis of epidemiological studies it has been proposed that cannabis use plays a causal role in the abuse of highly addictive drugs (Gateway Hypothesis). However, epidemiological studies are intrinsically unable to provide evidence of causality. Experimental studies can provide this evidence but they are feasible only in animal models and to date such evidence is lacking. In view of the importance of genetic factors in drug abuse, we investigated the influence of adolescent cannabis exposure on adult heroin reinforcement in two inbred rat strains differentially vulnerable to drugs of abuse, addiction prone Lewis (LEW) and addiction resistant Fischer 344 (F344) strains. Male LEW and F344 rats aged six weeks were exposed to increasing Δ9-tetrahydrocannabinol (THC) doses, twice a day for 3 days (2, 4, 8 mg/kg, i.p.). At adulthood they were allowed to self-administer heroin (0.025 mg/kg) under both Fixed- (FR) and Progressive- (PR) ratio schedules of responding. Following extinction, responding was reinstated by drug-cues and/or by heroin priming. THC pre-exposure increased responding for heroin and heroin intake under FR-3 and FR-5 as well as PR protocols and increased breaking point in PR schedules in LEW but not F344 rats. Drug cues and heroin priming reinstated responding in LEW and F344, but THC pre-exposure increased reinstatement by priming in LEW rats and by cues in F344 rats. These observations show that in genetically predisposed individuals, adolescent cannabis exposure increases heroin reinforcing properties, thus providing a mechanism for a causal role of adolescent cannabis use in heroin abuse.


Subject(s)
Analgesics, Opioid/administration & dosage , Behavior, Addictive/genetics , Behavior, Addictive/psychology , Cannabis , Heroin/administration & dosage , Reinforcement, Psychology , Age Factors , Animals , Behavior, Addictive/chemically induced , Dronabinol/administration & dosage , Male , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Rats, Transgenic , Self Administration
17.
Neurochem Res ; 45(4): 891-901, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31981057

ABSTRACT

Drug addiction can be described as a chronic and relapsing brain disease. Behavioral sensitization is believed to share similar mechanisms with relapse. Our previous studies have demonstrated that ifenprodil could attenuate methamphetamine (METH)-induced behavioral sensitization. However, the mechanism underlying this process has not been fully investigated. Protein phosphatase 2A (PP2A) is a conserved serine/threonine protein phosphatase that has been linked to many neurological diseases; however, there are few reports about PP2A in the context of drug addiction. In this study, we measured the level of phosphorylated (p-) GluN2B (Serine; Ser 1303), PP2A/B (a regulatory subunit of PP2A), and PP2A/C (a catalytic subunit of PP2A) in different brain regions such as the prefrontal cortex (PFc), nucleus accumbens (NAc), dorsal striatum (DS), and hippocampus (Hip). We also used ifenprodil, a selective antagonist of GluN2B to clarify the relationship between GluN2B and PP2A. The results showed that METH increased the level of p-GluN2B (Ser 1303) and PP2A/B in the DS and ifenprodil blocked this increase. We further examined the interaction between PP2A/B and PP2A/C in the DS and found that METH treatment increased the interaction between PP2A/B and PP2A/C, which was also blocked by ifenprodil. Then, we explored the pathway downstream of PP2A in the DS and found that p-AKT (Threonine; Thr 308) but not p-AKT (Ser 473) was dephosphorylated by PP2A. Taken together, these results indicated that the GluN2B-PP2A-AKT cascade was involved in METH-induced behavioral sensitization.


Subject(s)
Behavior, Addictive/prevention & control , Corpus Striatum/drug effects , Piperidines/therapeutic use , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Behavior, Addictive/chemically induced , Corpus Striatum/metabolism , Male , Methamphetamine , Mice, Inbred C57BL , Signal Transduction/drug effects , Substance-Related Disorders/drug therapy
18.
Psychopharmacology (Berl) ; 237(5): 1359-1369, 2020 May.
Article in English | MEDLINE | ID: mdl-31996940

ABSTRACT

RATIONALE: The nicotine delivery rate is a key feature of tobacco product design, yet there have been limited human studies examining the effects of nicotine as a function of delivery rate. OBJECTIVE: We developed an intravenous nicotine infusion protocol to evaluate differential effects of nicotine delivery rate on subjective drug effects, smoking urges, abstinence symptoms, heart rate, and blood pressure. METHODS: Eighteen non-treatment seeking, overnight abstinent male and female smokers (18 to 30 years old), who smoked ≥ 5 cigarettes per day for the past year completed four sessions, in which they were randomly assigned to a saline infusion, or a 1 mg per 70-kg body weight dose of nicotine delivered over 1, 5, or 10 min at rates of 0.24, 0.048, or 0.024 µg/kg/s, respectively. RESULTS: Smoking urges, as assessed by the Brief Questionnaire of Smoking Urges, were reduced relative to placebo for the 1- and 5-min infusion, but not the 10-min infusion. Although the 1- and 5-min infusions reduced smoking urges to a similar extent, the 1-min infusion induced a greater heart rate and blood pressure increase. Changes to subjective drug effects, heart rate, and blood pressure delineate the differential effects of nicotine delivery rate for these outcomes. CONCLUSIONS: We have characterized the delivery rate-response curve for a nicotine dose that is roughly the amount of nicotine (~ 1 mg) delivered by smoking a standard tobacco cigarette. Our findings reinforce the importance of nicotine delivery rate when evaluating the potential effects of nicotine from tobacco products.


Subject(s)
Blood Pressure/drug effects , Drug Delivery Systems/methods , Heart Rate/drug effects , Nicotine/administration & dosage , Smokers/psychology , Tobacco Smoking/psychology , Adult , Behavior, Addictive/chemically induced , Behavior, Addictive/psychology , Blood Pressure/physiology , Female , Heart Rate/physiology , Humans , Infusions, Intravenous , Male , Tobacco Smoking/physiopathology , Young Adult
19.
J Psychopharmacol ; 34(3): 336-347, 2020 03.
Article in English | MEDLINE | ID: mdl-31475622

ABSTRACT

BACKGROUND: There has been growing interest in using oxytocin as a pharmacotherapy for psychiatric disorders, including substance use disorder. Limited data exist regarding oxytocin's reinforcing efficacy, which is a necessary consideration for novel pharmacotherapies, especially in substance-using populations. AIMS: This study aimed to determine the potential reinforcing effects of intranasally administered oxytocin by assessing behavioral economic demand and subjective effects. METHODS: Healthy adults (n = 23) participated in a double-blind, repeated-measures, laboratory study wherein they received intranasal oxytocin (40 IU) or placebo in a randomized order across two sessions. Participants completed drug purchasing tasks at the conclusion of both sessions. Throughout both sessions, subjective and physiological effects were assessed. RESULTS: Demand-curve analysis of purchasing tasks revealed greater median purchasing for oxytocin relative to placebo. Physiological and subjective effects did not significantly differ between oxytocin and placebo. However, a nonsignificant trend was observed for moderately greater drug liking for oxytocin relative to placebo. There was a significant, positive correlation between the difference in drug liking (between oxytocin and placebo) and the difference in lowest-price purchasing (between oxytocin and placebo). CONCLUSIONS: These data suggest the potential for limited reinforcing and abuse-related subjective effects of intranasal oxytocin. Given the small sample, the greater drug liking of oxytocin compared to placebo, and the positive relation between demand and drug liking, it is possible that oxytocin may produce reinforcing effects in some participants. Therefore, additional studies of oxytocin reinforcement are warranted.


Subject(s)
Behavior, Addictive/chemically induced , Oxytocin/pharmacology , Reinforcement, Psychology , Administration, Intranasal , Adult , Consumer Behavior , Double-Blind Method , Female , Humans , Male , Oxytocin/adverse effects , Young Adult
20.
Addict Biol ; 25(6): e12843, 2020 11.
Article in English | MEDLINE | ID: mdl-31733097

ABSTRACT

Glutamatergic plasticity in the nucleus accumbens core (NAcore) is a key neuronal process in appetitive learning and contributes to pathologies such as drug addiction. Understanding how this plasticity factors into cannabis addiction and relapse has been hampered by the lack of a rodent model of cannabis self-administration. We used intravenous self-administration of two constituents of cannabis, Δ9 -tetrahydrocannabinol (THC) and cannabidiol (CBD) to examine how contingent cannabis use and cue-induced cannabinoid-seeking alters glutamatergic neurotransmission and synaptic plasticity in NAcore. NMDA receptor (NMDAR)-dependent long-term depression (LTD) in the NAcore was lost after cannabinoid, but not sucrose self-administration. Surprisingly, when rats underwent cue-induced cannabinoid seeking, LTD was restored. Loss of LTD was accompanied by desensitization of cannabinoid receptor 1 (CB1R). CB1R are positioned to regulate synaptic plasticity by being expressed on glutamatergic terminals and negatively regulating presynaptic excitability and glutamate release. Supporting this possibility, LTD was restored by promoting CB1R signaling with the CB1 positive allosteric modulator GAT211. These data implicate NAcore CB1R as critical regulators of metaplasticity induced by cannabis self-administration and the cues predicting cannabis availability.


Subject(s)
Cannabidiol/pharmacology , Cannabinoids/pharmacology , Dronabinol/pharmacology , Long-Term Synaptic Depression/drug effects , Receptor, Cannabinoid, CB1/agonists , Receptors, N-Methyl-D-Aspartate/physiology , Allosteric Regulation/drug effects , Animals , Behavior, Addictive/chemically induced , Cannabidiol/administration & dosage , Cannabinoid Receptor Agonists/administration & dosage , Cannabinoid Receptor Agonists/pharmacology , Cannabinoids/administration & dosage , Dronabinol/administration & dosage , Glutamic Acid/metabolism , Indoles/administration & dosage , Indoles/pharmacology , Long-Term Synaptic Depression/physiology , Male , Nucleus Accumbens/drug effects , Rats , Rats, Sprague-Dawley , Self Administration , Synaptic Transmission/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...