1
|
Carbone MG, Maremmani I. Chronic Cocaine Use and Parkinson's Disease: An Interpretative Model. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:1105. [PMID: 39200714 PMCID: PMC11354226 DOI: 10.3390/ijerph21081105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/18/2024] [Accepted: 08/16/2024] [Indexed: 09/02/2024]
Abstract
Over the years, the growing "epidemic" spread of cocaine use represents a crucial public health and social problem worldwide. According to the 2023 World Drug Report, 0.4% of the world's population aged 15 to 64 report using cocaine; this number corresponds to approximately 24.6 million cocaine users worldwide and approximately 1 million subjects with cocaine use disorder (CUD). While we specifically know the short-term side effects induced by cocaine, unfortunately, we currently do not have exhaustive information about the medium/long-term side effects of the substance on the body. The scientific literature progressively highlights that the chronic use of cocaine is related to an increase in cardio- and cerebrovascular risk and probably to a greater incidence of psychomotor symptoms and neurodegenerative processes. Several studies have highlighted an increased risk of antipsychotic-induced extrapyramidal symptoms (EPSs) in patients with psychotic spectrum disorders comorbid with psychostimulant abuse. EPSs include movement dysfunction such as dystonia, akathisia, tardive dyskinesia, and characteristic symptoms of Parkinsonism such as rigidity, bradykinesia, and tremor. In the present paper, we propose a model of interpretation of the neurobiological mechanisms underlying the hypothesized increased vulnerability in chronic cocaine abusers to neurodegenerative disorders with psychomotor symptoms. Specifically, we supposed that the chronic administration of cocaine produces significant neurobiological changes, causing a complex dysregulation of various neurotransmitter systems, mainly affecting subcortical structures and the dopaminergic pathways. We believe that a better understanding of these cellular and molecular mechanisms involved in cocaine-induced neuropsychotoxicity may have helpful clinical implications and provide targets for therapeutic intervention.
Collapse
Affiliation(s)
- Manuel Glauco Carbone
- Division of Psychiatry, Department of Medicine and Surgery, University of Insubria, Viale Luigi Borri 57, 21100 Varese, Italy;
- VP Dole Research Group, G. De Lisio Institute of Behavioural Sciences, Via di Pratale 3, 56121 Pisa, Italy
- Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
| | - Icro Maremmani
- VP Dole Research Group, G. De Lisio Institute of Behavioural Sciences, Via di Pratale 3, 56121 Pisa, Italy
- Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
- Addiction Research Methods Institute, World Federation for the Treatment of Opioid Dependence, 225 Varick Street, Suite 402, New York, NY 10014, USA
| |
Collapse
|
2
|
Chauhan H, Carruthers N, Stemmer P, Schneider BP, Moszczynska A. Neurotoxic Methamphetamine Doses Alter CDCel-1 Levels and Its Interaction with Vesicular Monoamine Transporter-2 in Rat Striatum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.21.604458. [PMID: 39091864 PMCID: PMC11291068 DOI: 10.1101/2024.07.21.604458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
In recent years, methamphetamine METH misuse in the US has been rapidly increasing and there is no FDA-approved pharmacotherapy for METH use disorder (MUD). In addition to being dependent on the drug, people with MUD develop a variety of neurological problems related to the toxicity of this drug. A variety of molecular mechanisms underlying METH neurotoxicity has been identified, including dysfunction of the neuroprotective protein parkin. However, it is not known whether parkin loss of function within striatal dopaminergic (DAergic) terminals translates into a decrease in DA storage capacity. This study examined the relationship between parkin, its substrate cell division cycle related-1 (CDCrel-1), and vesicular monoamine transporter-2 (VMAT2) in METH neurotoxicity in male Sprague Dawley rats. To also assess individual differences in response to METH's neurotoxic effects, a large group of rats was treated with binge METH or saline and sacrificed 1h or 24h later. This study is the first to show that binge METH alters the levels and subcellular localization of CDCrel-1 and that CDCrel-1 interacts with VMAT2 and increases its levels at the plasma membrane. Furthermore, we found wide individual differences in the responses of measured indices to METH. Proteomic analysis of VMAT-2-associated proteins revealed upregulation of several proteins involved in the exocytosis/endocytosis cycle. The results suggest that at 1h after METH binge, DAergic neurons are engaged in counteracting METH-induced toxic effects, including oxidative stress- and hyperthermia-induced inhibition of synaptic vesicle cycling, with the responses varying between individual rats. Studying CDCrel-1, VMAT2, and other proteins in large groups of outbred rats can help define individual genetic and molecular differences in responses to METH neurotoxicity which, in turn, will aid treating humans suffering from METH use disorder and its neurological consequences.
Collapse
Affiliation(s)
- Heli Chauhan
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI, USA 48201
| | - Nick Carruthers
- Institute of Environmental Health Sciences and Proteomics Core Facility, 540 East Canfield Ave., Detroit, MI 48202
| | - Paul Stemmer
- Institute of Environmental Health Sciences and Proteomics Core Facility, 540 East Canfield Ave., Detroit, MI 48202
| | - Bernard P. Schneider
- Brain Mind Institute École Polytechnique Fédérale de Lausanne School of Life Sciences, Ch. Des Mines, 9, CH-1202 Geneve, Switzerland
| | - Anna Moszczynska
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Ave, Detroit, MI, USA 48201
| |
Collapse
|
3
|
Sarparast A, Thomas K, Malcolm B, Stauffer CS. Drug-drug interactions between psychiatric medications and MDMA or psilocybin: a systematic review. Psychopharmacology (Berl) 2022; 239:1945-1976. [PMID: 35253070 PMCID: PMC9177763 DOI: 10.1007/s00213-022-06083-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/03/2022] [Indexed: 12/13/2022]
Abstract
RATIONALE & OBJECTIVES ± 3,4-Methylenedioxymethamphetamine (MDMA) and psilocybin are currently moving through the US Food and Drug Administration's phased drug development process for psychiatric treatment indications: posttraumatic stress disorder and depression, respectively. The current standard of care for these disorders involves treatment with psychiatric medications (e.g., selective serotonin reuptake inhibitors), so it will be important to understand drug-drug interactions between MDMA or psilocybin and psychiatric medications. METHODS In accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we queried the MEDLINE database via PubMed for publications of human studies in English spanning between the first synthesis of psilocybin (1958) and December 2020. We used 163 search terms containing 22 psychiatric medication classes, 135 specific psychiatric medications, and 6 terms describing MDMA or psilocybin. RESULTS Forty publications were included in our systematic review: 26 reporting outcomes from randomized controlled studies with healthy adults, 3 epidemiologic studies, and 11 case reports. Publications of studies describe interactions between MDMA (N = 24) or psilocybin (N = 5) and medications from several psychiatric drug classes: adrenergic agents, antipsychotics, anxiolytics, mood stabilizers, NMDA antagonists, psychostimulants, and several classes of antidepressants. We focus our results on pharmacodynamic, physiological, and subjective outcomes of drug-drug interactions. CONCLUSIONS As MDMA and psilocybin continue to move through the FDA drug development process, this systematic review offers a compilation of existing research on psychiatric drug-drug interactions with MDMA or psilocybin.
Collapse
Affiliation(s)
- Aryan Sarparast
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Kelan Thomas
- College of Pharmacy, Touro University California, Vallejo, CA, 94592, USA
| | | | - Christopher S Stauffer
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, 97239, USA.
- Department of Mental Health, VA Portland Health Care System, Portland, OR, 97239, USA.
| |
Collapse
|
4
|
SheikhBahaei S, Farhan M, Maguire GA. Improvement of stuttering after administration of methylphenidate - a case report. PERSONALIZED MEDICINE IN PSYCHIATRY 2022; 31-32. [PMID: 35237733 PMCID: PMC8881904 DOI: 10.1016/j.pmip.2022.100092] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
5
|
McDougall SA, Robinson JAM, Gleason DC, Cotter LL. Reciprocal cross-sensitization between cocaine and RU 24969 in male and female preweanling rats. Pharmacol Biochem Behav 2021; 209:173265. [PMID: 34437872 DOI: 10.1016/j.pbb.2021.173265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/19/2021] [Accepted: 08/20/2021] [Indexed: 10/20/2022]
Abstract
Neuronal adaptations involving dopaminergic, glutamatergic, and serotonergic neurotransmitter systems are responsible for behavioral sensitization. Because of common underlying mechanisms, cross-sensitization between compounds of different drug classes can be observed. The purpose of the present study was to determine whether a one- or four-day pretreatment regimen of RU 24969 (a 5-HT1A/1B receptor agonist) would reciprocally cross-sensitize with cocaine or methamphetamine in male and female preweanling rats. Rats were pretreated with RU 24969 (0 or 5 mg/kg) for 4 days (PD 17-20) and then challenged with cocaine (10 or 20 mg/kg) or methamphetamine (1 or 2 mg/kg) on PD 22. Reciprocal cross-sensitization was also assessed (i.e., rats were pretreated with psychostimulants and tested with RU 24969). In a follow-up experiment, the ability of RU 24969 and cocaine to reciprocally cross-sensitize was assessed using a one-day pretreatment regimen. Reciprocal cross-sensitization between cocaine and RU 24969 was evident in preweanling rats, whereas methamphetamine and RU 24969 did not cross-sensitize. When a one-trial pretreatment regimen was used, cross-sensitization was only detected when rats were pretreated with RU 24969 and tested with cocaine, but not the reverse. In sum, the present results show that the nonselective 5-HT1A/1B receptor agonist RU 24969 cross-sensitizes with cocaine, but not methamphetamine, in preweanling rats. This dichotomy may be a function of cocaine having a greater affinity for the serotonin transporter than methamphetamine.
Collapse
Affiliation(s)
- Sanders A McDougall
- Department of Psychology, 5500 University Parkway, California State University, San Bernardino, CA 92407, USA.
| | - Jasmine A M Robinson
- Department of Psychology, 5500 University Parkway, California State University, San Bernardino, CA 92407, USA
| | - Devon C Gleason
- Department of Psychology, 5500 University Parkway, California State University, San Bernardino, CA 92407, USA
| | - Laura L Cotter
- Department of Psychology, 5500 University Parkway, California State University, San Bernardino, CA 92407, USA
| |
Collapse
|
6
|
Docherty JR, Alsufyani HA. Pharmacology of Drugs Used as Stimulants. J Clin Pharmacol 2021; 61 Suppl 2:S53-S69. [PMID: 34396557 DOI: 10.1002/jcph.1918] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/22/2021] [Indexed: 12/21/2022]
Abstract
Psychostimulant, cardiovascular, and temperature actions of stimulants involve adrenergic (norepinephrine), dopaminergic (dopamine), and serotonergic (serotonin) pathways. Stimulants such as amphetamine, 3,4-methylenedioxymethamphetamine (MDMA), or mephedrone can act on the neuronal membrane monoamine transporters NET, DAT, and SERT and/or the vesicular monoamine transporter 2 to inhibit reuptake of neurotransmitter or cause release by reverse transport. Stimulants may have additional effects involving pre- and postsynaptic/junctional receptors for norepinephrine, dopamine, and serotonin and other receptors. As a result, stimulants may have a wide range of possible actions. Agents with cocaine or MDMA-like actions can induce serious and potentially fatal adverse events via thermodysregulatory, cardiovascular, or other mechanisms. MDMA-like stimulants may cause hyperthermia that can be life threathening. Recreational users of stimulants should be aware of the dangers of hyperthermia in a rave/club environment.
Collapse
Affiliation(s)
| | - Hadeel A Alsufyani
- Department of Physiology, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
7
|
Grønnestad R, Johanson SM, Müller MHB, Schlenk D, Tanabe P, Krøkje Å, Jaspers VLB, Jenssen BM, Ræder EM, Lyche JL, Shi Q, Arukwe A. Effects of an environmentally relevant PFAS mixture on dopamine and steroid hormone levels in exposed mice. Toxicol Appl Pharmacol 2021; 428:115670. [PMID: 34371090 DOI: 10.1016/j.taap.2021.115670] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022]
Abstract
In the present study, we investigated the dopaminergic and steroid hormone systems of A/J mice fed environmentally relevant concentrations of a perfluoroalkyl substance (PFAS) mixture over a period of 10 weeks. The PFAS mixture was chosen based on measured PFAS concentrations in earthworms at a Norwegian skiing area (Trondheim) and consisted of eight different PFAS. Dietary exposure to PFAS led to lower total brain dopamine (DA) concentrations in male mice, as compared to control. On the transcript level, brain tyrosine hydroxylase (th) of PFAS exposed males was reduced, compared to the control group. No significant differences were observed on the transcript levels of enzymes responsible for DA metabolism, namely - monoamine oxidase (maoa and maob) and catechol-O methyltransferase (comt). We detected increased transcript level for DA receptor 2 (dr2) in PFAS exposed females, while expression of DA receptor 1 (dr1), DA transporter (dat) and vesicular monoamine transporter (vmat) were not affected by PFAS exposure. Regarding the steroid hormones, plasma and muscle testosterone (T), 11-ketotestosterone (11-KT) and 17β-estradiol (E2) levels, as well as transcripts for estrogen receptors (esr1 and esr2), gonadotropin releasing hormone (gnrh) and aromatase (cyp19) were unaltered by the PFAS treatment. These results indicate that exposure to PFAS doses, comparable to previous observation in earthworms at a Norwegian skiing area, may alter the dopaminergic system of mice with overt consequences for health, general physiology, cognitive behavior, reproduction and metabolism.
Collapse
Affiliation(s)
- Randi Grønnestad
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Silje Modahl Johanson
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Mette H B Müller
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, Oslo, Norway; Department of Paraclinical Sciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Daniel Schlenk
- Department of Environmental Sciences, University of California, Riverside, California, USA
| | - Philip Tanabe
- Department of Environmental Sciences, University of California, Riverside, California, USA
| | - Åse Krøkje
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Veerle L B Jaspers
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Bjørn Munro Jenssen
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biosciences, Aarhus University, Roskilde, Denmark
| | - Erik M Ræder
- Department of Paraclinical Sciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Jan L Lyche
- Department of Paraclinical Sciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Qingyang Shi
- Department of Environmental Sciences, University of California, Riverside, California, USA
| | - Augustine Arukwe
- Department of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
8
|
Sakai C. [Pharmacological properties and clinical effects of the ADHD drug, Lisdexamfetamine (Vyvanse ® capsules 20 mg and 30 mg)]. Nihon Yakurigaku Zasshi 2020; 155:413-425. [PMID: 33132261 DOI: 10.1254/fpj.20033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Lisdexamfetamine dimesylate (hereinafter referred to as "lisdexamfetamine"; brand name, Vyvanse®), was developed for the treatment of attention-deficit/hyperactivity disorder (ADHD). This drug, which is classified as a central nervous system (CNS) stimulant for once-daily oral administration, received marketing approval in March 2019 and was launched in December 2019 in Japan. Lisdexamfetamine is a prodrug that is hydrolyzed to its active form d-amphetamine in the blood following oral administration. Pharmacologically, d-amphetamine competitively inhibits the dopamine transporter (DAT) and the noradrenaline transporter (NAT) to increase dopamine (DA) and noradrenaline (NA) concentrations in the synaptic cleft. In addition to inhibiting the reuptake of DA and NA, d-amphetamine has also an effect in promoting the release of these neurotransmitters by being taken up into neuronal cells and then acting on the vesicular monoamine transporter. The mechanisms of action by which d-amphetamine exerts a therapeutic effect on ADHD may be based on the above-described effects. Clinical studies conducted in Japan and overseas have demonstrated the efficacy of lisdexamfetamine over placebo in the treatment of pediatric ADHD patients. The most of the adverse events with a higher incidence than in the placebo were mild, and long-term administration of the drug was not associated with an increase in the incidence of adverse events or the rate of treatment discontinuation. Lisdexamfetamine, which is designated as raw material for stimulants and therefore requires strict distribution control in Japan, is expected to be effective in the treatment of ADHD patients with inadequate responses to existing therapeutic agents.
Collapse
Affiliation(s)
- Chika Sakai
- Medical Affairs Department, Shionogi & Co., Ltd
| |
Collapse
|
9
|
Crawford CA, Teran A, Ramirez GI, Katz CG, Mohd-Yusof A, Eaton SE, Real V, McDougall SA. Age-dependent effects of dopamine receptor inactivation on cocaine-induced behaviors in male rats: Evidence of dorsal striatal D2 receptor supersensitivity. J Neurosci Res 2019; 97:1546-1558. [PMID: 31304635 DOI: 10.1002/jnr.24491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/09/2019] [Accepted: 06/14/2019] [Indexed: 12/21/2022]
Abstract
N-ethoxycarbonyl-2-ethoxy-1,2-dihydroquinoline (EEDQ), which irreversibly inactivates dopamine (DA) receptors, causes pronounced age-dependent behavioral effects in rats. For example, EEDQ either augments or does not affect the DA agonist-induced locomotor activity of preweanling rats while attenuating the locomotion of adolescent and adult rats. The twofold purpose of this study was to determine whether EEDQ would: (a) potentiate or attenuate the cocaine-induced locomotor activity of preweanling, adolescent, and adult rats; and (b) alter the sensitivity of surviving D2 receptors. Rats were treated with vehicle or EEDQ (2.5 or 7.5 mg/kg) on postnatal day (PD) 17, PD 39, and PD 84. In the behavioral experiments, saline- or cocaine-induced locomotion was assessed 24 hr later. In the biochemical experiments, dorsal striatal samples were taken 24 hr after vehicle or EEDQ treatment and later assayed for NPA-stimulated GTPγS receptor binding, G protein-coupled receptor kinase 6 (GRK6), and β-arrestin-2 (ARRB2). GTPγS binding is a direct measure of ligand-induced G protein activation, while GRK6 and ARRB2 modulate the internalization and desensitization of D2 receptors. Results showed that EEDQ potentiated the locomotor activity of preweanling rats, while attenuating the locomotion of older rats. NPA-stimulated GTPγS binding was elevated in EEDQ-treated preweanling rats, relative to adults, indicating enhanced functional coupling between the G protein and receptor. EEDQ also reduced ARRB2 levels in all age groups, which is indicative of increased D2 receptor sensitivity. In sum, the present results support the hypothesis that D2 receptor supersensitivity is a critical factor mediating the locomotor potentiating effects of EEDQ in cocaine-treated preweanling rats.
Collapse
Affiliation(s)
- Cynthia A Crawford
- Department of Psychology, California State University, San Bernardino, California
| | - Angie Teran
- Department of Psychology, California State University, San Bernardino, California
| | - Goretti I Ramirez
- Department of Psychology, California State University, San Bernardino, California
| | - Caitlin G Katz
- Department of Psychology, California State University, San Bernardino, California
| | - Alena Mohd-Yusof
- Department of Psychology, California State University, San Bernardino, California
| | - Shannon E Eaton
- Department of Psychology, California State University, San Bernardino, California
- Department of Psychology, University of Kentucky, Lexington, Kentucky
| | - Vanessa Real
- Department of Psychology, California State University, San Bernardino, California
| | - Sanders A McDougall
- Department of Psychology, California State University, San Bernardino, California
| |
Collapse
|
10
|
Cholanians AB, Phan AV, Lau SS, Monks TJ. Concurrent Inhibition of Vesicular Monoamine Transporter 2 Does Not Protect Against 3,4-Methylenedioxymethamphetamine (Ecstasy) Induced Neurotoxicity. Toxicol Sci 2019; 170:157-166. [PMID: 30923810 DOI: 10.1093/toxsci/kfz085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
3, 4-Methylenedioxymethamphetamine (MDMA) is a hallucinogenic amphetamine derivative. The acute effects of MDMA are hyperthermia, hyperactivity, and behavioral changes, followed by long-term serotonergic neurotoxicity in rats and primates. However, the underlying mechanisms of MDMA neurotoxicity remain elusive. We reported that pretreatment of rats with Ro 4-1284, a reversible inhibitor of the vesicular monoamine transporter 2 (VMAT2), reduced MDMA-induced hyperactivity in rats, abolished the hyperthermic response, and the long-term neurotoxicity. Current studies focused on the effects of co- and/or postinhibition of VMAT2 on the acute and chronic effects of MDMA and on the dose-response relationship between MDMA-induced elevations in body temperature and subsequent reductions in indolamine concentrations. Sprague Dawley rats were treated with MDMA (20, 25, or 27.5 mg/kg sc), and either co- and/or posttreatment with the VMAT2 inhibitor (10 mg/kg ip). Rats simultaneously treated with Ro 4-1284 and MDMA exhibited a more rapid increase in body temperature compared to just MDMA. However, the duration of the elevated body temperature was significantly shortened (approximately 3 h vs approximately 8 h, respectively). A similar body temperature response was observed in rats posttreated (7 h after MDMA) with Ro 4-1284. Despite decreases in the area under the curve (Δtemp X time) of body temperature caused by Ro 4-1284, there were no significant differences in the degree of indolamine depletion between any of the MDMA-treated groups. The results suggest that the neuroprotective effects of VMAT2 inhibition is likely due to the indirect monoamine depleting effects of the Ro 4-1284 pretreatment, rather than by the direct inhibition of VMAT2 function.
Collapse
Affiliation(s)
- Aram B Cholanians
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721
| | - Andy V Phan
- Department of Radiation Oncology, School of Medicine, University of Colorado Denver, Denver, Colorado 80045
| | - Serrine S Lau
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan 48201
| | - Terrence J Monks
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan 48201
| |
Collapse
|
11
|
Naglich AC, Brown ES, Adinoff B. Systematic review of preclinical, clinical, and post-marketing evidence of bupropion misuse potential. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2019; 45:341-354. [PMID: 30601027 DOI: 10.1080/00952990.2018.1545023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Background: Bupropion is a substituted cathinone compound widely used as a first line or add-on treatment for depression, smoking cessation, and more recently in combination with naltrexone for weight loss. As abuse of synthetic cathinone compounds has received more attention in recent years, concern about the misuse potential of bupropion has grown as well. Objectives: We review bupropion pharmacology and assessments of misuse potential including preclinical evidence, human studies, and post-marketing surveillance of bupropion misuse. Methods: This review reports the results of a systematic review of publications evaluating the potential for bupropion to be misused. Publications were identified using PubMed and Medline through Ovid® as well as iterative bibliographic searches. A summary of data from informal sources of information including substance-user experience from online forum entries is included. Results: Preclinical evidence demonstrates some potential for misuse based on psychomotor, discrimination, self-administration, and conditioned place preference tasks. However, this potential is less than that of commonly misused stimulants. Studies in human populations similarly indicate that bupropion shares interoceptive effects with other stimulants, but lacks some key reinforcing effects of other stimulants. In the real-world setting, misuse of bupropion occurs, but is uncommon. Adverse effects of bupropion misuse are frequently cited as significant barriers to obtaining any desired interoceptive effect. Conclusions: While bupropion demonstrates some potential for misuse, pharmacological differences from other structurally-related stimulants limit bupropion's reinforcing effects. Without additional data indicating susceptibility of specific populations to bupropion misuse, there is no empirical data suggesting a need to modify bupropion prescribing patterns.
Collapse
Affiliation(s)
- Andrew C Naglich
- a Department of Mental Health , VA North Texas Healthcare System , Dallas , TX , USA
| | - E Sherwood Brown
- b Department of Psychiatry , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Bryon Adinoff
- c University of Colorado School of Medicine , Denver , CO , USA
| |
Collapse
|
12
|
Mohd-Yusof A, Veliz A, Rudberg KN, Stone MJ, Gonzalez AE, McDougall SA. Effects of D2 or combined D1/D2 receptor antagonism on the methamphetamine-induced one-trial and multi-trial behavioral sensitization of preweanling rats. Psychopharmacology (Berl) 2016; 233:893-903. [PMID: 26650612 PMCID: PMC4752886 DOI: 10.1007/s00213-015-4170-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/22/2015] [Indexed: 11/30/2022]
Abstract
RATIONALE There is suggestive evidence that the neural mechanisms mediating one-trial and multi-trial behavioral sensitization differ, especially when the effects of various classes of dopamine (DA) agonists are examined. OBJECTIVE The purpose of the present study was to determine the role of the D2 receptor for the induction of one-trial and multi-trial methamphetamine sensitization in preweanling rats. METHODS In a series of experiments, rats were injected with saline or raclopride (a selective D2 receptor antagonist), either alone or in combination with SCH23390 (a selective D1 receptor antagonist), 15 min prior to treatment with the indirect DA agonist methamphetamine. Acute control groups were given two injections of saline. This pretreatment regimen occurred on either postnatal days (PD) 13-16 (multi-trial) or PD 16 (one-trial). On PD 17, rats were challenged with methamphetamine and locomotor sensitization was determined. RESULTS Blockade of D2 or D1/D2 receptors reduced or prevented, respectively, the induction of multi-trial methamphetamine sensitization in young rats, while the same manipulations had minimal effects on one-trial behavioral sensitization. CONCLUSIONS DA antagonist treatment differentially affected the methamphetamine-induced sensitized responding of preweanling rats depending on whether a one-trial or multi-trial procedure was used. The basis for this effect is uncertain, but there was some evidence that repeated DA antagonist treatment caused nonspecific changes that produced a weakened sensitized response. Importantly, DA antagonist treatment did not prevent the one-trial behavioral sensitization of preweanling rats. The latter result brings into question whether DA receptor stimulation is necessary for the induction of psychostimulant-induced behavioral sensitization during early ontogeny.
Collapse
Affiliation(s)
- Alena Mohd-Yusof
- Department of Psychology, 5500 University Parkway, California State University, San Bernardino, CA, 92407, USA
| | - Ana Veliz
- Department of Psychology, 5500 University Parkway, California State University, San Bernardino, CA, 92407, USA
| | - Krista N Rudberg
- Department of Psychology, 5500 University Parkway, California State University, San Bernardino, CA, 92407, USA
| | - Michelle J Stone
- Department of Psychology, 5500 University Parkway, California State University, San Bernardino, CA, 92407, USA
| | - Ashley E Gonzalez
- Department of Psychology, 5500 University Parkway, California State University, San Bernardino, CA, 92407, USA
- Present address: A.E. Gonzalez, Neurosciences Program, Stanford University School of Medicine, 1215 Welch Road Modular B, #42, Stanford, CA, 94305-5400, USA
| | - Sanders A McDougall
- Department of Psychology, 5500 University Parkway, California State University, San Bernardino, CA, 92407, USA.
| |
Collapse
|
13
|
Somkuwar SS, Staples MC, Fannon MJ, Ghofranian A, Mandyam CD. Evaluating Exercise as a Therapeutic Intervention for Methamphetamine Addiction-Like Behavior. Brain Plast 2015; 1:63-81. [PMID: 29765835 PMCID: PMC5928557 DOI: 10.3233/bpl-150007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The need for effective treatments for addiction and dependence to the illicit stimulant methamphetamine in primary care settings is increasing, yet no effective medications have been FDA approved to reduce dependence [1]. This is partially attributed to the complex and dynamic neurobiology underlying the various stages of addiction [2]. Therapeutic strategies to treat methamphetamine addiction, particularly the relapse stage of addiction, could revolutionize methamphetamine addiction treatment. In this context, preclinical studies demonstrate that voluntary exercise (sustained physical activity) could be used as an intervention to reduce methamphetamine addiction. Therefore, it appears that methamphetamine disrupts normal functioning in the brain and this disruption is prevented or reduced by engaging in exercise. This review discusses animal models of methamphetamine addiction and sustained physical activity and the interactions between exercise and methamphetamine behaviors. The review highlights how methamphetamine and exercise affect neuronal plasticity and neurotoxicity in the adult mammalian striatum, hippocampus, and prefrontal cortex, and presents the emerging mechanisms of exercise in attenuating intake and in preventing relapse to methamphetamine seeking in preclinical models of methamphetamine addiction.
Collapse
Affiliation(s)
- Sucharita S Somkuwar
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Miranda C Staples
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - McKenzie J Fannon
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Atoosa Ghofranian
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Chitra D Mandyam
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
14
|
Seol I, Lim D, Lee J, Yang H. Comparative studies on behavioral neurochemical effects of N,α-diethylphenethylamine and methamphetamine in rats. Forensic Toxicol 2014. [DOI: 10.1007/s11419-014-0254-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
15
|
Jones CG, Yang PB, Wilcox VT, Dafny N. Amphetamine Alters the Circadian Locomotor Activity Pattern of Adult WKY Female Rats. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/jbbs.2014.45022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
16
|
McDougall SA, Nuqui CM, Quiroz AT, Martinez CM. Early ontogeny of D-amphetamine-induced one-trial behavioral sensitization. Pharmacol Biochem Behav 2013; 104:154-62. [PMID: 23360956 DOI: 10.1016/j.pbb.2013.01.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Revised: 11/30/2012] [Accepted: 01/16/2013] [Indexed: 11/19/2022]
Abstract
The early ontogeny of D-amphetamine-induced one-trial behavioral sensitization was characterized using male and female preweanling and preadolescent rats. In Experiment 1, rats were injected with saline or D-amphetamine (1, 4, or 8mg/kg) in activity chambers or the home cage on postnatal day (PD) 12, PD 16, PD 20, or PD 24. One day later, rats were challenged with either 0.5 or 2mg/kg D-amphetamine and distance traveled was measured in activity chambers for 120min. In Experiment 2, saline or D-amphetamine was administered in activity chambers on PD 24, while a challenge injection of D-amphetamine (0.25-4mg/kg) was given on PD 25. At younger ages (PD 13 and PD 17), a strong sensitized response was evident on the test day regardless of whether rats were pretreated with D-amphetamine (4 or 8mg/kg) before being placed in the activity chamber or 30min after being returned to the home cage. Rats did not display D-amphetamine-induced behavioral sensitization on PD 21, nor was context-dependent sensitization apparent on PD 25 even when a broad dose range of D-amphetamine was used. When low doses of D-amphetamine were administered on the pretreatment and test days (1 and 0.5mg/kg, respectively), sensitized responding was not evident at any age. In summary, D-amphetamine-induced one-trial behavioral sensitization was only apparent within a narrow developmental window during early ontogeny. This ontogenetic pattern of sensitized responding is similar to the one produced by methamphetamine and distinct from the pattern produced by cocaine. The unique sensitization profiles resulting from repeated D-amphetamine and cocaine treatment may be a consequence of their different mechanisms of action.
Collapse
Affiliation(s)
- Sanders A McDougall
- Department of Psychology, 5500 University Parkway, California State University, San Bernardino, CA 92407, USA.
| | | | | | | |
Collapse
|
17
|
Carvalho M, Carmo H, Costa VM, Capela JP, Pontes H, Remião F, Carvalho F, Bastos MDL. Toxicity of amphetamines: an update. Arch Toxicol 2012; 86:1167-231. [PMID: 22392347 DOI: 10.1007/s00204-012-0815-5] [Citation(s) in RCA: 261] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 02/02/2012] [Indexed: 01/06/2023]
Abstract
Amphetamines represent a class of psychotropic compounds, widely abused for their stimulant, euphoric, anorectic, and, in some cases, emphathogenic, entactogenic, and hallucinogenic properties. These compounds derive from the β-phenylethylamine core structure and are kinetically and dynamically characterized by easily crossing the blood-brain barrier, to resist brain biotransformation and to release monoamine neurotransmitters from nerve endings. Although amphetamines are widely acknowledged as synthetic drugs, of which amphetamine, methamphetamine, and 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) are well-known examples, humans have used natural amphetamines for several millenniums, through the consumption of amphetamines produced in plants, namely cathinone (khat), obtained from the plant Catha edulis and ephedrine, obtained from various plants in the genus Ephedra. More recently, a wave of new amphetamines has emerged in the market, mainly constituted of cathinone derivatives, including mephedrone, methylone, methedrone, and buthylone, among others. Although intoxications by amphetamines continue to be common causes of emergency department and hospital admissions, it is frequent to find the sophism that amphetamine derivatives, namely those appearing more recently, are relatively safe. However, human intoxications by these drugs are increasingly being reported, with similar patterns compared to those previously seen with classical amphetamines. That is not surprising, considering the similar structures and mechanisms of action among the different amphetamines, conferring similar toxicokinetic and toxicological profiles to these compounds. The aim of the present review is to give an insight into the pharmacokinetics, general mechanisms of biological and toxicological actions, and the main target organs for the toxicity of amphetamines. Although there is still scarce knowledge from novel amphetamines to draw mechanistic insights, the long-studied classical amphetamines-amphetamine itself, as well as methamphetamine and MDMA, provide plenty of data that may be useful to predict toxicological outcome to improvident abusers and are for that reason the main focus of this review.
Collapse
Affiliation(s)
- Márcia Carvalho
- REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Blakely RD, Edwards RH. Vesicular and plasma membrane transporters for neurotransmitters. Cold Spring Harb Perspect Biol 2012; 4:a005595. [PMID: 22199021 PMCID: PMC3281572 DOI: 10.1101/cshperspect.a005595] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The regulated exocytosis that mediates chemical signaling at synapses requires mechanisms to coordinate the immediate response to stimulation with the recycling needed to sustain release. Two general classes of transporter contribute to release, one located on synaptic vesicles that loads them with transmitter, and a second at the plasma membrane that both terminates signaling and serves to recycle transmitter for subsequent rounds of release. Originally identified as the target of psychoactive drugs, these transport systems have important roles in transmitter release, but we are only beginning to understand their contribution to synaptic transmission, plasticity, behavior, and disease. Recent work has started to provide a structural basis for their activity, to characterize their trafficking and potential for regulation. The results indicate that far from the passive target of psychoactive drugs, neurotransmitter transporters undergo regulation that contributes to synaptic plasticity.
Collapse
Affiliation(s)
- Randy D Blakely
- Department of Pharmacology and Psychiatry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-8548, USA
| | | |
Collapse
|
19
|
Loewinger GC, Beckert MV, Tejeda HA, Cheer JF. Methamphetamine-induced dopamine terminal deficits in the nucleus accumbens are exacerbated by reward-associated cues and attenuated by CB1 receptor antagonism. Neuropharmacology 2012; 62:2192-201. [PMID: 22306525 DOI: 10.1016/j.neuropharm.2012.01.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 12/24/2011] [Accepted: 01/18/2012] [Indexed: 01/28/2023]
Abstract
Methamphetamine (METH) exposure is primarily associated with deleterious effects to dopaminergic neurons. While several studies have implicated the endocannabinoid system in METH's locomotor, rewarding and neurochemical effects, a role for this signaling system in METH's effects on dopamine terminal dynamics has not been elucidated. Given that CB1 receptor blockade reduces the acute potentiation of phasic extracellular dopamine release from other psychomotor stimulant drugs and that the degree of acute METH-induced increases in extracellular dopamine levels is related to the severity of dopamine depletion, we predicted that pretreatment with the CB1 receptor antagonist rimonabant would reduce METH-induced alterations at dopamine terminals. Furthermore, we hypothesized that administration of METH in environments where reward associated-cues were present would potentiate METH's acute effects on dopamine release in the nucleus accumbens and exacerbate changes in dopamine terminal activity. Fast-scan cyclic voltammetry was used to measure electrically-evoked dopamine release in the nucleus accumbens and revealed markers of compromised dopamine terminal integrity nine days after a single dose of METH. These were exacerbated in animals that received METH in the presence of reward-associated cues, and attenuated in rimonabant-pretreated animals. While these deficits in dopamine dynamics were associated with reduced operant responding on days following METH administration in animals treated with only METH, rimonabant-pretreated animals exhibited levels of operant responding comparable to control. Moreover, dopamine release correlated significantly with changes in lever pressing behavior that occurred on days following METH administration. Together these data suggest that the endocannabinoid system is involved in the subsecond dopaminergic response to METH.
Collapse
Affiliation(s)
- Gabriel C Loewinger
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn Street, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
20
|
Afanador L, Yarosh H, Wang J, Ali SF, Angulo JA. Contrasting Effects of the Neuropeptides Substance P, Somatostatin, and Neuropeptide Y on the Methamphetamine-Induced Production of Striatal Nitric Oxide in Mice. ACTA ACUST UNITED AC 2012; 1. [PMID: 25383232 DOI: 10.4303/jdar/235604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Several laboratories have shown that methamphetamine (METH) neurotoxicity is associated with increases of nitric oxide (NO) production in striatal tissue and blockade of NO production protects from METH. Because substance P modulates NO production, we tested the hypothesis that intrinsic striatal neuropeptides such as somatostatin and neuropeptide Y (NPY) modulate striatal NO production in the presence of METH. To that end, METH (30 mg/kg, IP) was injected into adult male mice alone or in combination with pharmacological agonists or antagonists of the neurokinin-1 (substance P), somatostatin or NPY receptors and 3-nitrotyrosine (an indirect index of NO production) was assessed utilizing HPLC or a histological method. Pre-treatment with the systemic neurokinin-1 receptor antagonist WIN-51,708 significantly attenuated the METH-induced production of striatal 3-NT measured at two hours post-METH. Conversely, intrastriatal injection of NPY1 or 2 receptor agonists inhibited the METH-induced production of striatal 3-NT. Similarly, intrastriatal infusion of the somatostatin receptor agonist octreotide attenuated the METH-induced striatal production of 3-NT. Taken together, our results suggest the hypothesis that the neuropeptide substance P is pro-damage while the neuropeptides somatostatin and NPY are anti-damage in the presence of METH by targeting the production of NO.
Collapse
Affiliation(s)
- Lauriaselle Afanador
- Department of Biological Sciences, Hunter College, 695 Park Avenue, NY 10021, USA ; The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Haley Yarosh
- Department of Biological Sciences, Hunter College, 695 Park Avenue, NY 10021, USA ; The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Jing Wang
- Department of Biological Sciences, Hunter College, 695 Park Avenue, NY 10021, USA ; The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Syed F Ali
- Neurochemistry Laboratory, Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR 72079, USA
| | - Jesus A Angulo
- Department of Biological Sciences, Hunter College, 695 Park Avenue, NY 10021, USA ; The Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| |
Collapse
|
21
|
Glajch KE, Fleming SM, Surmeier DJ, Osten P. Sensorimotor assessment of the unilateral 6-hydroxydopamine mouse model of Parkinson's disease. Behav Brain Res 2011; 230:309-16. [PMID: 22178078 DOI: 10.1016/j.bbr.2011.12.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 11/30/2011] [Accepted: 12/04/2011] [Indexed: 12/30/2022]
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disorder, is characterized by marked impairments in motor function caused by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNc). Animal models of PD have traditionally been based on toxins, such as 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), that selectively lesion dopaminergic neurons. Motor impairments from 6-OHDA lesions of SNc neurons are well characterized in rats, but much less work has been done in mice. In this study, we compare the effectiveness of a series of drug-free behavioral tests in assessing sensorimotor impairments in the unilateral 6-OHDA mouse model, including six tests used for the first time in this PD mouse model (the automated treadmill "DigiGait" test, the challenging beam test, the adhesive removal test, the pole test, the adjusting steps test, and the test of spontaneous activity) and two tests used previously in 6-OHDA-lesioned mice (the limb-use asymmetry "cylinder" test and the manual gait test). We demonstrate that the limb-use asymmetry, challenging beam, pole, adjusting steps, and spontaneous activity tests are all highly robust assays for detecting sensorimotor impairments in the 6-OHDA mouse model. We also discuss the use of the behavioral tests for specific experimental objectives, such as simple screening for well-lesioned mice in studies of PD cellular pathophysiology or comprehensive behavioral analysis in preclinical therapeutic testing using a battery of sensorimotor tests.
Collapse
Affiliation(s)
- Kelly E Glajch
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60610, USA.
| | | | | | | |
Collapse
|
22
|
Crooks PA, Zheng G, Vartak AP, Culver JP, Zheng F, Horton DB, Dwoskin LP. Design, synthesis and interaction at the vesicular monoamine transporter-2 of lobeline analogs: potential pharmacotherapies for the treatment of psychostimulant abuse. Curr Top Med Chem 2011; 11:1103-27. [PMID: 21050177 DOI: 10.2174/156802611795371332] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Accepted: 08/30/2010] [Indexed: 11/22/2022]
Abstract
The vesicular monoamine transporter-2 (VMAT2) is considered as a new target for the development of novel therapeutics to treat psychostimulant abuse. Current information on the structure, function and role of VMAT2 in psychostimulant abuse are presented. Lobeline, the major alkaloidal constituent of Lobelia inflata, interacts with nicotinic receptors and with VMAT2. Numerous studies have shown that lobeline inhibits both the neurochemical and behavioral effects of amphetamine in rodents, and behavioral studies demonstrate that lobeline has potential as a pharmacotherapy for psychostimulant abuse. Systematic structural modification of the lobeline molecule is described with the aim of improving selectivity and affinity for VMAT2 over neuronal nicotinic acetylcholine receptors and other neurotransmitter transporters. This has led to the discovery of more potent and selective ligands for VMAT2. In addition, a computational neural network analysis of the affinity of these lobeline analogs for VMAT2 has been carried out, which provides computational models that have predictive value in the rational design of VMAT2 ligands and is also useful in identifying drug candidates from virtual libraries for subsequent synthesis and evaluation.
Collapse
Affiliation(s)
- Peter A Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, 40536-0082, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Siviy SM, Crawford CA, Akopian G, Walsh JP. Dysfunctional play and dopamine physiology in the Fischer 344 rat. Behav Brain Res 2011; 220:294-304. [PMID: 21335036 DOI: 10.1016/j.bbr.2011.02.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 02/03/2011] [Accepted: 02/06/2011] [Indexed: 02/01/2023]
Abstract
Juvenile Fischer 344 rats are known to be less playful than other inbred strains, although the neurobiological substrate(s) responsible for this phenotype is uncertain. In the present study, Fischer 344 rats were compared to the commonly used outbred Sprague-Dawley strain on several behavioral and physiological parameters in order to ascertain whether the lack of play may be related to compromised activity of brain dopamine (DA) systems. As expected, Fischer 344 rats were far less playful than Sprague-Dawley rats, with Fischer 344 rats less likely to initiate playful contacts with a playful partner and less likely to respond playfully to these contacts. We also found that Fischer 344 rats showed less of a startle response and greater pre-pulse inhibition (PPI), especially at higher pre-pulse intensities. The increase in PPI seen in the Fischer 344 rat could be due to reduced DA modulation of sensorimotor gating and neurochemical measures were consistent with Fischer 344 rats releasing less DA than Sprague-Dawley rats. Fast scan cyclic voltammetry (FSCV) revealed Fischer 344 rats had less evoked DA release in dorsal and ventral striatal brain slices and high-performance liquid chromatography revealed Fischer 344 rats to have less DA turnover in the striatum and prefrontal cortex. We also found DA-dependent forms of cortical plasticity were deficient in the striatum and prefrontal cortex of the Fischer 344 rat. Taken together, these data indicate that deficits in play and enhanced PPI of Fischer 344 rats may be due to reduced DA modulation of corticostriatal and mesolimbic/mesocortical circuits critical to the execution of these behaviors.
Collapse
Affiliation(s)
- Stephen M Siviy
- Department of Psychology, Gettysburg College, Gettysburg, PA 17325, USA.
| | | | | | | |
Collapse
|
24
|
Melatonin attenuates the amphetamine-induced decrease in vesicular monoamine transporter-2 expression in postnatal rat striatum. Neurosci Lett 2011; 488:154-7. [DOI: 10.1016/j.neulet.2010.11.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 11/05/2010] [Accepted: 11/05/2010] [Indexed: 11/18/2022]
|
25
|
Wang J, Angulo JA. Synergism between methamphetamine and the neuropeptide substance P on the production of nitric oxide in the striatum of mice. Brain Res 2010; 1369:131-9. [PMID: 21075091 DOI: 10.1016/j.brainres.2010.11.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 11/04/2010] [Accepted: 11/05/2010] [Indexed: 11/27/2022]
Abstract
Our laboratory has been investigating the participation of striatal neurokinin-1 receptors in the methamphetamine (METH)-induced loss of striatal neurons. Signaling through these receptors exacerbates the METH-induced striatal apoptosis. METH induces the synthesis of nitric oxide (NO) and the latter has been linked to the activation of neurodegenerative cascades. In the present study, we assessed the role of the neurokinin-1 receptor in the production of striatal 3-nitrotyrosine (3-NT) and l-citrulline (indirect indices of NO production). To that end, we injected male mice with a bolus of METH (30 mg/kg, ip) and visualized striatal neuronal nitric oxide synthase (NOS)-positive cells by immunohistochemistry and protein levels by Western blot. The expression of neuronal NOS or protein levels at 2, 4 and 8 hours post-METH was unchanged. Next, we assessed 3-NT and l-citrulline by immunohistochemistry. At 4 hours post-METH, striatal 3-NT and l-citrulline levels were increased 30- and 5-fold, respectively, relative to controls and the selective neurokinin-1 receptor antagonist WIN-51,708 attenuated these increases. Intrastriatal infusion of the neurokinin-1 receptor agonist GR-73632 induced striatal 3-NT production that was attenuated with systemic injection of WIN-51,708 or 7-nitroindazole (7-NI, an inhibitor of neuronal NOS). Moreover, infusion of calmidazolium (calmodulin inhibitor) with GR-73632 prevented the production of 3-NT. These data are consistent with the hypothesis that METH-induced production of NO is modulated by the striatal neurokinin-1 receptors and that this receptor may participate in the biochemical activation of neuronal NOS.
Collapse
Affiliation(s)
- Jing Wang
- Hunter College of the City University of New York, Department of Biological Sciences, 695 Park Avenue, New York, NY 10021, USA
| | | |
Collapse
|
26
|
Seger D. Cocaine, metamfetamine, and MDMA abuse: the role and clinical importance of neuroadaptation. Clin Toxicol (Phila) 2010; 48:695-708. [DOI: 10.3109/15563650.2010.516263] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
27
|
Chronic low dose Adderall XR down-regulates cfos expression in infantile and prepubertal rat striatum and cortex. Neuroscience 2010; 169:1901-12. [PMID: 20600661 DOI: 10.1016/j.neuroscience.2010.06.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 05/31/2010] [Accepted: 06/12/2010] [Indexed: 11/23/2022]
Abstract
We previously reported that treatment of prepubertal male rats with low, injected or oral, doses of methylphenidate stimulated cfos, fosB and arc expression in many areas of the developing brain. In the present study our objective was to determine whether the widely prescribed psychostimulant Adderall XR (ADD) exerted similar effects in infantile and prepubertal rat brain. We report here, for the first time, that low threshold doses of oral ADD, an extended-release mixture of amphetamine salts, now routinely used for the treatment of Attention Deficit Hyperactivity Disorder (ADHD), also increased cfos expression in infantile (postnatal day 10; PD10) and prepubertal (PD24) rat brain. These threshold doses were correlated with blood levels of amphetamine determined by liquid chromatography-mass spectrometry. Moreover, we observed that chronic treatment with oral ADD (1.6 mg/kg; x 14 days) not only significantly down-regulated cfos expression following a final challenge dose of ADD in prepubertal (PD24) rat striatum and cortex, quantified in terms of FOS immunoreactivity (FOS-ir), but did so at a daily dose that was without effect with methylphenidate (MPH); that is a much higher oral dose of MPH (7.5 mg/kg; x 14 days) failed to induce down-regulation of cfos expression. Similar experiments in infantile rats (PD10), but using a threshold injected dose of ADD (1.25 mg/kg sc) also significantly reduced striatal and cingulate cortical FOS-ir. An additional finding in the prepubertal rats was that oral ADD-induced FOS-ir was observed in the cerebral cortex following doses lower than the threshold dose necessary to increase FOS-ir in the striatum. This was not the case in the PD10 rats. In conclusion, our efforts to calibrate biological responses, such as immediate early gene expression, to clinically relevant blood levels of stimulants confirmed that expression of cfos is very sensitive to repeated low doses of Adderall XR. It is now feasible to examine whether other genes are also affected in these young rats and if the changes we report are reversible. The implications of such studies should be relevant to the putative effects of psychostimulant treatment of very young children.
Collapse
|
28
|
Grygoruk A, Fei H, Daniels RW, Miller BR, Diantonio A, Krantz DE. A tyrosine-based motif localizes a Drosophila vesicular transporter to synaptic vesicles in vivo. J Biol Chem 2010; 285:6867-78. [PMID: 20053989 DOI: 10.1074/jbc.m109.073064] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vesicular neurotransmitter transporters must localize to synaptic vesicles (SVs) to allow regulated neurotransmitter release at the synapse. However, the signals required to localize vesicular proteins to SVs in vivo remain unclear. To address this question we have tested the effects of mutating proposed trafficking domains in Drosophila orthologs of the vesicular monoamine and glutamate transporters, DVMAT-A and DVGLUT. We show that a tyrosine-based motif (YXXY) is important both for DVMAT-A internalization from the cell surface in vitro, and localization to SVs in vivo. In contrast, DVGLUT deletion mutants that lack a putative C-terminal trafficking domain show more modest defects in both internalization in vitro and trafficking to SVs in vivo. Our data show for the first time that mutation of a specific trafficking motif can disrupt localization to SVs in vivo and suggest possible differences in the sorting of VMATs versus VGLUTs to SVs at the synapse.
Collapse
Affiliation(s)
- Anna Grygoruk
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Hatos Center for Neuropharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California 90095-1761, USA
| | | | | | | | | | | |
Collapse
|
29
|
Scheidweiler KB, Ladenheim B, Cadet JL, Huestis MA. Mice lacking multidrug resistance protein 1a show altered dopaminergic responses to methylenedioxymethamphetamine (MDMA) in striatum. Neurotox Res 2009; 18:200-9. [PMID: 19851718 DOI: 10.1007/s12640-009-9124-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 08/04/2009] [Accepted: 10/07/2009] [Indexed: 01/16/2023]
Abstract
Multidrug resistance protein 1a (MDR1a) potentiated methylenedioxymethamphetamine (MDMA)-induced decreases of dopamine (DA) and dopamine transport protein in mouse brain one week after MDMA administration. In the present study, we examined if mdr1a wild-type (mdr1a +/+) and knock-out (mdr1a -/-) mice differentially handle the acute effects of MDMA on the nigrostriatal DA system 0-24 h following a single drug injection. 3-way ANOVA revealed significant 2-way interactions of strain x time (F (5,152) = 32.4, P < 0.001) and strain x dose (F (3,152) = 25.8, P < 0.001) on 3,4-dihydroxyphenylacetic acid (DOPAC)/DA ratios in mdr1a +/+ and -/- mice. 0.3-3 h after 10 mg/kg MDMA, DOPAC/DA ratios were increased in mdr1a +/+ mice, but decreased 0.3-1 h after MDMA in mdr1a -/- mice. Twenty-four hours after 10 mg/kg MDMA, DOPAC/DA ratios were increased 600% in mdr1a +/+ mice compared to saline-treated control mice, while in mdr1a -/- mice DOPAC/DA ratios were unchanged. Striatal MDMA and its metabolite, methylenedioxyamphetamine, concentrations by gas chromatography-mass spectrometry were similar in both strains 0.3-4 h after MDMA, discounting the role of MDR1a-facilitated MDMA transport in observed inter-strain differences. Increased DOPAC/DA turnover in mdr1a +/+ mice following MDMA is consistent with the previous report that MDMA neurotoxicity is increased in mdr1a +/+ mice. Increased DA turnover via monoamine oxidase in mdr1a +/+ vs -/- mice might increase exposure to neurotoxic reactive oxygen species.
Collapse
Affiliation(s)
- Karl B Scheidweiler
- Chemistry and Drug Metabolism, Intramural Research Program, National Institutes of Health, Biomedical Research Center, 251 Bayview Boulevard Suite 200, Room 05A-721, Baltimore, MD 21224, USA
| | | | | | | |
Collapse
|
30
|
Superoxide production after acute and chronic treatment with methylphenidate in young and adult rats. Neurosci Lett 2009; 465:95-8. [PMID: 19716398 DOI: 10.1016/j.neulet.2009.08.060] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 08/24/2009] [Accepted: 08/24/2009] [Indexed: 11/21/2022]
Abstract
The prescription of methylphenidate (MPH) has dramatically increased in this decade for attention deficit hyperactivity disorder (ADHD) treatment. The action mechanism of MPH is not completely understood and studies have been demonstrated that MPH can lead to neurochemical adaptations. Superoxide radical anion is not very reactive per se. However, severe species derived from superoxide radical anion mediate most of its toxicity. In this study, the superoxide level in submitochondrial particles was evaluated in response to treatment with MPH in the age-dependent manner in rats. MPH was administrated acutely or chronically at doses of 1, 2 or 10 mg/kg i.p. The results showed that the acute administration of MPH in all doses in young rats increased the production of superoxide in the cerebellum and only in the high dose (10mg/kg) in the hippocampus, while chronic treatment had no effect. However, acute treatment in adult rats had no effect on production of superoxide, but chronic treatment decreased the production of superoxide in the cerebellum at the lower doses. Our data suggest that the MPH treatment can influence on production of superoxide in some brain areas, but this effect depends on age of animals and treatment regime with MPH.
Collapse
|
31
|
Narendran R, Martinez D. Cocaine abuse and sensitization of striatal dopamine transmission: a critical review of the preclinical and clinical imaging literature. Synapse 2009; 62:851-69. [PMID: 18720516 DOI: 10.1002/syn.20566] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Much effort has been devoted in the preclinical addiction literature to understanding the phenomenon of sensitization, an enhanced dopaminergic response in the nucleus accumbens that occurs after repeated exposure to psychostimulant drugs. Although sensitization has been reported in preclinical studies, studies of sensitization in humans measuring behavioral and physiological responses have been mixed and inconclusive. However, imaging studies with positron emission tomography (PET) and single photon emission computed tomography (SPECT) using a stimulant challenge to induce dopamine (DA) release provide a unique opportunity to probe DA transmission in cocaine dependent human subjects. In contrast to the basic science literature that predicted sensitization, three independent cohorts have shown a blunted DA response, or the opposite of sensitization, in human cocaine dependent subjects. This article reviews the methodological differences between the preclinical and clinical PET studies that have investigated DA sensitization in order to address the discrepancy between the human and animal literature.
Collapse
Affiliation(s)
- Rajesh Narendran
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15232, USA.
| | | |
Collapse
|
32
|
Capela JP, Carmo H, Remião F, Bastos ML, Meisel A, Carvalho F. Molecular and Cellular Mechanisms of Ecstasy-Induced Neurotoxicity: An Overview. Mol Neurobiol 2009; 39:210-71. [DOI: 10.1007/s12035-009-8064-1] [Citation(s) in RCA: 210] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 02/27/2009] [Indexed: 11/29/2022]
|
33
|
Pharmacology of stimulants prohibited by the World Anti-Doping Agency (WADA). Br J Pharmacol 2008; 154:606-22. [PMID: 18500382 DOI: 10.1038/bjp.2008.124] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
This review examines the pharmacology of stimulants prohibited by the World Anti-Doping Agency (WADA). Stimulants that increase alertness/reduce fatigue or activate the cardiovascular system can include drugs like ephedrine available in many over-the-counter medicines. Others such as amphetamines, cocaine and hallucinogenic drugs, available on prescription or illegally, can modify mood. A total of 62 stimulants (61 chemical entities) are listed in the WADA List, prohibited in competition. Athletes may have stimulants in their body for one of three main reasons: inadvertent consumption in a propriety medicine; deliberate consumption for misuse as a recreational drug and deliberate consumption to enhance performance. The majority of stimulants on the list act on the monoaminergic systems: adrenergic (sympathetic, transmitter noradrenaline), dopaminergic (transmitter dopamine) and serotonergic (transmitter serotonin, 5-HT). Sympathomimetic describes agents, which mimic sympathetic responses, and dopaminomimetic and serotoninomimetic can be used to describe actions on the dopamine and serotonin systems. However, many agents act to mimic more than one of these monoamines, so that a collective term of monoaminomimetic may be useful. Monoaminomimietic actions of stimulants can include blockade of re-uptake of neurotransmitter, indirect release of neurotransmitter, direct activation of monoaminergic receptors. Many of the stimulants are amphetamines or amphetamine derivatives, including agents with abuse potential as recreational drugs. A number of agents are metabolized to amphetamine or metamphetamine. In addition to the monoaminomimetic agents, a small number of agents with different modes of action are on the list. A number of commonly used stimulants are not considered as Prohibited Substances.
Collapse
|
34
|
Kuperstein F, Eilam R, Yavin E. Altered expression of key dopaminergic regulatory proteins in the postnatal brain following perinatal n-3 fatty acid dietary deficiency. J Neurochem 2008; 106:662-71. [DOI: 10.1111/j.1471-4159.2008.05418.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
35
|
Chu PW, Seferian KS, Birdsall E, Truong JG, Riordan JA, Metcalf CS, Hanson GR, Fleckenstein AE. Differential regional effects of methamphetamine on dopamine transport. Eur J Pharmacol 2008; 590:105-10. [PMID: 18599036 DOI: 10.1016/j.ejphar.2008.05.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 04/18/2008] [Accepted: 05/20/2008] [Indexed: 10/22/2022]
Abstract
Multiple high-dose methamphetamine administrations cause long-lasting (>1 week) deficits in striatal dopaminergic neuronal function. This stimulant likewise causes rapid (within 1 h) and persistent (at least 48 h) decreases in activities of striatal: 1) dopamine transporters, as assessed in synaptosomes; and 2) vesicular monoamine transporter -2 (VMAT-2), as assessed in a non-membrane-associated (referred to herein as cytoplasmic) vesicular subcellular fraction. Importantly, not all brain areas are vulnerable to methamphetamine-induced long-lasting deficits. Similarly, the present study indicates that methamphetamine exerts differential acute effects on monoaminergic transporters according to brain region. In particular, results revealed that in the nucleus accumbens, methamphetamine rapidly, but reversibly (within 24 h), decreased plasmalemmal dopamine transporter function, without effect on plasmalemmal dopamine transporter immunoreactivity. Methamphetamine also rapidly and reversibly (within 48 h) decreased cytoplasmic VMAT-2 function in this region, with relatively little effect on cytoplasmic VMAT-2 immunoreactivity. In contrast, methamphetamine did not alter either dopamine transporter or VMAT-2 activity in the hypothalamus. Noteworthy, the nucleus accumbens and hypothalamus did not display the persistent long-lasting striatal dopamine depletions caused by the stimulant. Taken together, these data suggest that deficits in plasmalemmal and vesicular monoamine transporter activity lasting greater than 24-48 h may be linked to the long-lasting dopaminergic deficits caused by methamphetamine and appear to be region specific.
Collapse
Affiliation(s)
- Pei-Wen Chu
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Kurling S, Kankaanpää A, Seppälä T. Sub-chronic nandrolone treatment modifies neurochemical and behavioral effects of amphetamine and 3,4-methylenedioxymethamphetamine (MDMA) in rats. Behav Brain Res 2008; 189:191-201. [DOI: 10.1016/j.bbr.2007.12.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Accepted: 12/28/2007] [Indexed: 11/29/2022]
|
37
|
Ravna AW, Sager G, Dahl SG, Sylte I. Membrane Transporters: Structure, Function and Targets for Drug Design. TOPICS IN MEDICINAL CHEMISTRY 2008. [DOI: 10.1007/7355_2008_023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
38
|
Chen MK, Kuwabara H, Zhou Y, Adams RJ, Brasić JR, McGlothan JL, Verina T, Burton NC, Alexander M, Kumar A, Wong DF, Guilarte TR. VMAT2 and dopamine neuron loss in a primate model of Parkinson's disease. J Neurochem 2007; 105:78-90. [PMID: 17988241 DOI: 10.1111/j.1471-4159.2007.05108.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We used positron emission tomography (PET) to measure the earliest change in dopaminergic synapses and glial cell markers in a chronic, low-dose MPTP non-human primate model of Parkinson's disease (PD). In vivo levels of dopamine transporters (DAT), vesicular monoamine transporter-type 2 (VMAT2), amphetamine-induced dopamine release (AMPH-DAR), D2-dopamine receptors (D2R) and translocator protein 18 kDa (TSPO) were measured longitudinally in the striatum of MPTP-treated animals. We report an early (2 months) decrease (46%) of striatal VMAT2 in asymptomatic MPTP animals that preceded changes in DAT, D2R, and AMPH-DAR and was associated with increased TSPO levels indicative of a glial response. Subsequent PET studies showed progressive loss of all pre-synaptic dopamine markers in the striatum with expression of parkinsonism. However, glial cell activation did not track disease progression. These findings indicate that decreased VMAT2 is a key pathogenic event that precedes nigrostriatal dopamine neuron degeneration. The loss of VMAT2 may result from an association with alpha-synuclein aggregation induced by oxidative stress. Disruption of dopamine sequestration by reducing VMAT2 is an early pathogenic event in the dopamine neuron degeneration that occurs in the MPTP non-human primate model of PD. Genetic or environmental factors that decrease VMAT2 function may be important determinants of PD.
Collapse
Affiliation(s)
- Ming-Kai Chen
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Changes in the response to release of a single synaptic vesicle have generally been attributed to postsynaptic modification of receptor sensitivity, but considerable evidence now demonstrates that alterations in vesicle filling also contribute to changes in quantal size. Receptors are not saturated at many synapses, and changes in the amount of transmitter per vesicle contribute to the physiological regulation of release. On the other hand, the presynaptic factors that determine quantal size remain poorly understood. Aside from regulation of the fusion pore, these mechanisms fall into two general categories: those that affect the accumulation of transmitter inside a vesicle and those that affect vesicle size. This review will summarize current understanding of the neurotransmitter cycle and indicate basic, unanswered questions about the presynaptic regulation of quantal size.
Collapse
Affiliation(s)
- Robert H Edwards
- Department of Neurology and Physiology, UCSF School of Medicine, San Francisco, CA 94158-2517, USA.
| |
Collapse
|
40
|
Ji J, McDermott JL, Dluzen DE. Sex differences in K+-evoked striatal dopamine output from superfused striatal tissue fragments of reserpine-treated CD-1 mice. J Neuroendocrinol 2007; 19:725-31. [PMID: 17680888 DOI: 10.1111/j.1365-2826.2007.01581.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Reserpine inhibits vesicular monoamine transporter-2 (VMAT-2) function and thereby impairs vesicular dopamine (DA) storage within nerve terminals. The present report compared the effects of reserpine treatment upon the striatal dopaminergic system in male and female mice as a means to assess potential sex differences in VMAT-2/DA storage function. After treatment with reserpine, male mice showed significantly greater striatal DA concentrations and K+ -evoked DA output from the striatum compared to females. By contrast, no statistically significant sex differences were observed in methamphetamine-evoked DA output in reserpine-treated mice. These results demonstrate a clear sex difference in the striatal dopaminergic responses to reserpine and suggest that females possess a more active VMAT-2/DA storage capacity, as indicated by the greater degree of deficits observed when VMAT-2/DA storage function is inhibited by reserpine. Such findings have important implications for understanding some of the bases for sex differences in neurotoxicity and neurodegeneration of the nigrostriatal dopaminergic system.
Collapse
Affiliation(s)
- J Ji
- Department of Anatomy, Northeastern Ohio Universities College of Medicine, 4209 State Route 44, Rootstown, OH 44272, USA
| | | | | |
Collapse
|
41
|
Howell LL, Kimmel HL. Monoamine transporters and psychostimulant addiction. Biochem Pharmacol 2007; 75:196-217. [PMID: 17825265 DOI: 10.1016/j.bcp.2007.08.003] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Revised: 07/25/2007] [Accepted: 08/02/2007] [Indexed: 12/27/2022]
Abstract
Psychostimulants are a broadly defined class of drugs that stimulate the central and peripheral nervous systems as their primary pharmacological effect. The abuse liability of psychostimulants is well established and represents a significant public health concern. An extensive literature documents the critical importance of monoamines (dopamine, serotonin and norepinephrine) in the behavioral pharmacology and addictive properties of psychostimulants. In particular, the dopamine transporter plays a primary role in the reinforcing and behavioral-stimulant effects of psychostimulants in animals and humans. Moreover, both serotonin and norepinephrine systems can reliably modulate the neurochemical and behavioral effects of psychostimulants. However, there is a growing body of evidence that highlights complex interactions among additional neurotransmitter systems. Cortical glutamatergic systems provide important regulation of dopamine function, and inhibitory amino acid gamma-aminobutyric acid (GABA) systems can modulate basal dopamine and glutamate release. Repeated exposure to psychostimulants can lead to robust and enduring changes in neurobiological substrates, including monoamines, and corresponding changes in sensitivity to acute drug effects on neurochemistry and behavior. Significant advances in the understanding of neurobiological mechanisms underlying psychostimulant abuse and dependence have guided pharmacological treatment strategies to improve clinical outcome. In particular, functional agonist treatments may be used effectively to stabilize monoamine neurochemistry, influence behavior and lead to long-term abstinence. However, additional clinical studies are required in order to identify safe and efficacious pharmacotherapies.
Collapse
Affiliation(s)
- Leonard L Howell
- Division of Neuroscience, Yerkes National Primate Research Center, Emory University, 954 Gatewood Road NE, Atlanta, GA 30329, USA.
| | | |
Collapse
|
42
|
Riddle EL, Hanson GR, Fleckenstein AE. Therapeutic doses of amphetamine and methylphenidate selectively redistribute the vesicular monoamine transporter-2. Eur J Pharmacol 2007; 571:25-8. [PMID: 17618619 PMCID: PMC2581712 DOI: 10.1016/j.ejphar.2007.05.044] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2007] [Revised: 05/17/2007] [Accepted: 05/22/2007] [Indexed: 11/22/2022]
Abstract
High-dose administration of psychostimulants traffics the vesicular monoamine transporter-2 (VMAT-2), as assessed by subcellular fractionation of rat striatal tissue. This study demonstrates that administration of low doses of amphetamine or methylphenidate differentially traffic VMAT-2 within nerve terminals, with effects similar to those observed after high-dose administration. Trafficking of vesicular glutamate, acetylcholine, or GABA transporters was not altered by high-or low-dose amphetamine or methylphenidate treatment. These data represent the first report that amphetamine redistributes VMAT-2 protein. In addition, these data demonstrate that the trafficking of VMAT-2 after amphetamine or methylphenidate is selective for monoaminergic neurons.
Collapse
Affiliation(s)
- Evan L Riddle
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, USA.
| | | | | |
Collapse
|
43
|
Fleckenstein AE, Volz TJ, Riddle EL, Gibb JW, Hanson GR. New insights into the mechanism of action of amphetamines. Annu Rev Pharmacol Toxicol 2007; 47:681-98. [PMID: 17209801 DOI: 10.1146/annurev.pharmtox.47.120505.105140] [Citation(s) in RCA: 482] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Amphetamine is a psychostimulant commonly used to treat several disorders, including attention deficit, narcolepsy, and obesity. Plasmalemmal and vesicular monoamine transporters, such as the neuronal dopamine transporter and the vesicular monoamine transporter-2, are two of its principal targets. This review focuses on new insights, obtained from both in vivo and in vitro studies, into the molecular mechanisms whereby amphetamine, and the closely related compounds methamphetamine and methylenedioxymethamphetamine, cause monoamine, and particularly dopamine, release. These mechanisms include amphetamine-induced exchange diffusion, reverse transport, and channel-like transport phenomena as well as the weak base properties of amphetamine. Additionally, amphetamine analogs may affect monoamine transporters through phosphorylation, transporter trafficking, and the production of reactive oxygen and nitrogen species. All of these mechanisms have potential implications for both amphetamine- and methamphetamine-induced neurotoxicity, as well as dopaminergic neurodegenerative diseases.
Collapse
Affiliation(s)
- Annette E Fleckenstein
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA.
| | | | | | | | | |
Collapse
|
44
|
Abstract
BACKGROUND Methamphetamine abuse has become an increasing problem in both the United States and globally with concomitant increases in adverse medical, social and environmental sequelae. Behavioral therapies have been used with some success to treat methamphetamine abusers and dependent individuals, but are not universally efficacious. Methamphetamine has a rich pharmacology that theoretically provides many opportunities for potential pharmacotherapeutic intervention. Nevertheless, there are no approved medications with an indication for treating methamphetamine abusers or addicts at this time. AIM To describe briefly how methamphetamine functions and affects function in brain and report how basic researchers and clinicians are attempting to exploit and exploiting this knowledge to discover and develop effective pharmacotherapies. RESULTS Scientifically based approaches to medications development by evaluating medications that limit brain exposure to methamphetamine; modulate methamphetamine effects at vesicular monoamine transporter-2 (VMAT-2); or affect dopaminergic, serotonergic, GABAergic, and/or glutamatergic brain pathways that participate in methamphetamine's reinforcing effects are presented. CONCLUSION The evidence supports the rationale that pharmacotherapies to decrease methamphetamine use, or reduce craving during abstinence may be developed from altering the pharmacokinetics and pharmacodynamics of methamphetamine or its effects on appetitive systems in the brain.
Collapse
Affiliation(s)
- Frank J Vocci
- Division of Pharmacotherapies and Medical Consequences of Drug Abuse, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | | |
Collapse
|
45
|
Volz TJ, Fleckenstein AE, Hanson GR. Methamphetamine-induced alterations in monoamine transport: implications for neurotoxicity, neuroprotection and treatment. Addiction 2007; 102 Suppl 1:44-8. [PMID: 17493052 DOI: 10.1111/j.1360-0443.2007.01771.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIM To review studies delineating the neurotoxic effects of methamphetamine on monoamine transport in dopaminergic neurons of the striatum and nucleus accumbens. METHOD The scope of this review includes the English language dopamine transporter and vesicular monoamine transporter-2 primary literature to April 2006 identified by Pubmed, Science Citation Index and SciFinder Scholar literature searches. RESULTS Changes in the function of the plasmalemmal dopamine transporter and the vesicular monoamine transporter-2 are key components of methamphetamine-induced persistent dopaminergic deficits. These deficits include persistent reductions in dopamine content, dopamine transporter density and tyrosine hydroxylase activity. The striatum is susceptible to these effects of methamphetamine while the nucleus accumbens is resistant. Differences in dopamine transporter density and activity, extracellular dopamine levels and antioxidant levels in these two brain regions may, in part, account for the resistance of the nucleus accumbens. CONCLUSION These findings concerning the nature of methamphetamine-induced changes in plasmalemmal and vesicular dopamine transport have very important implications for drug targets and for understanding the etiology of dopaminergic neurodegenerative processes, such as those associated with methamphetamine addiction and Parkinson's disease.
Collapse
Affiliation(s)
- Trent J Volz
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | | | | |
Collapse
|
46
|
Geracitano R, Federici M, Bernardi G, Mercuri NB. On the effects of psychostimulants, antidepressants, and the antiparkinsonian drug levodopa on dopamine neurons. Ann N Y Acad Sci 2007; 1074:320-9. [PMID: 17105928 DOI: 10.1196/annals.1369.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The dopaminergic system constitutes the principal target of many psychostimulants, antidepressant, and antiparkinsonian drugs. The effects caused by these compounds are partly associated with an increased dopamine (DA) levels within the terminal areas of DA neurons and in the ventral midbrain. Therefore, several substances of abuse, antidepressants, and endogenous compounds (levodopa and trace amines [TAs]) regulate the activity of DA cells by activating D2 autoreceptors located on the terminals, soma, and dendrites. Considering our past and recent experimental studies on this issue, here we will briefly reexamine the mechanisms of action of several psychoactive drugs on DA neurons. In particular, we propose three different modalities by which the mesencephalic DA neurons can be regulated by drugs: amphetamine/TAs-like, cocaine-like, and levodopa-like. We, therefore, discuss the potential therapeutic and addictive properties of the psychoactive substances.
Collapse
Affiliation(s)
- Raffaella Geracitano
- Experimental Neurology, Laboratory - C.E.R.C.- Fondazione S. Lucia I.R.C.C.S., Via del Fosso di Fiorano, 65-00143 Rome, Italy
| | | | | | | |
Collapse
|
47
|
Nutt DJ, Fone K, Asherson P, Bramble D, Hill P, Matthews K, Morris KA, Santosh P, Sonuga-Barke E, Taylor E, Weiss M, Young S. Evidence-based guidelines for management of attention-deficit/hyperactivity disorder in adolescents in transition to adult services and in adults: recommendations from the British Association for Psychopharmacology. J Psychopharmacol 2007; 21:10-41. [PMID: 17092962 DOI: 10.1177/0269881106073219] [Citation(s) in RCA: 185] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is an established diagnosis in children, associated with a large body of evidence on the benefits of treatment. Adolescents with ADHD are now leaving children's services often with no readily identifiable adult service to support them, which presents problems as local pharmacy regulations often preclude the prescription of stimulant drugs by general practitioners (GPs). In addition, adults with ADHD symptoms are now starting to present to primary care and psychiatry services requesting assessment and treatment. For these reasons, the British Association for Psychopharmacology (BAP) thought it timely to hold a consensus conference to review the body of evidence on childhood ADHD and the growing literature on ADHD in older age groups. Much of this initial guidance on managing ADHD in adolescents in transition and in adults is based on expert opinion derived from childhood evidence. We hope that, by the time these guidelines are updated, much evidence will be available to address the many directions for future research that are detailed here.
Collapse
Affiliation(s)
- D J Nutt
- Psychopharmacology Unit, University of Bristol, Bristol, and Bethlem Royal Hospital, Kent, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Volz TJ, Hanson GR, Fleckenstein AE. The role of the plasmalemmal dopamine and vesicular monoamine transporters in methamphetamine‐induced dopaminergic deficits. J Neurochem 2006; 101:883-8. [PMID: 17250674 DOI: 10.1111/j.1471-4159.2006.04419.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Amphetamine (AMPH) and methamphetamine (METH) are members of a collection of phenethylamine psychostimulants that are commonly referred to collectively as "amphetamines." Amphetamines exert their effects, in part, by affecting neuronal dopamine transport. This review thus focuses on the effects of AMPH and METH on the plasmalemmal dopamine transporter and the vesicular monoamine transporter-2 in animal models with a particular emphasis on how these effects, which may vary for the different stereoisomers, contribute to persistent dopaminergic deficits.
Collapse
Affiliation(s)
- Trent J Volz
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | | | | |
Collapse
|
49
|
Oades RD. Function and dysfunction of monoamine interactions in children and adolescents with AD/HD. EXS 2006; 98:207-44. [PMID: 17019890 DOI: 10.1007/978-3-7643-7772-4_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Robert D Oades
- Biopsychology Research Group, University Clinic for Child and Adolescent Psychiatry, Virchowstr 174, 45147 Essen, Germany.
| |
Collapse
|
50
|
Zheng G, Dwoskin LP, Crooks PA. Vesicular monoamine transporter 2: role as a novel target for drug development. AAPS JOURNAL 2006; 8:E682-92. [PMID: 17233532 PMCID: PMC2751365 DOI: 10.1208/aapsj080478] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the central nervous system, vesicular monoamine transporter 2 (VMAT2) is the only transporter that moves cytoplasmic dopamine (DA) into synaptic vesicles for storage and subsequent exocytotic release. Pharmacologically enhancing DA sequestration by VMAT2, and thus preventing the oxidation of DA in the cytoplasm, may be a strategy for treating diseases such as Parkinson's disease. VMAT2 may also be a novel target for the development of treatments for psychostimulant abuse. This review summarizes the possible role of VMAT2 as a therapeutic target, VMAT2 ligands reported in the literature, and the structure-activity relationship of these ligands, including tetrabenazine analogs, ketanserin analogs, lobeline analogs, and 3-amine-2-phenylpropene analogs. The molecular structure of VMAT2 and its relevance to ligand binding are briefly discussed.
Collapse
Affiliation(s)
- Guangrong Zheng
- College of Pharmacy, University of Kentucky, Department of Pharmaceutical Sciences, 907 Rose Street, Room 501B, 40536-0082 Lexington, KY
| | - Linda P. Dwoskin
- College of Pharmacy, University of Kentucky, Department of Pharmaceutical Sciences, 907 Rose Street, Room 501B, 40536-0082 Lexington, KY
| | - Peter A. Crooks
- College of Pharmacy, University of Kentucky, Department of Pharmaceutical Sciences, 907 Rose Street, Room 501B, 40536-0082 Lexington, KY
| |
Collapse
|