1
|
Hiranita T, Li SM, Katz JL. Effects of Dual Inhibition at Dopamine Transporter and σ Receptors in the Discriminative-Stimulus Effects of Cocaine in Male Rats. J Pharmacol Exp Ther 2024; 391:308-316. [PMID: 39179413 PMCID: PMC11493437 DOI: 10.1124/jpet.124.002239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
Previous studies demonstrated that sigma receptor (σR) antagonists alone fail to alter cocaine self-administration despite blocking various other effects of cocaine. However, σR antagonists when combined with dopamine transporter (DAT) inhibitors substantially decrease cocaine self-administration. To better understand the effects of this combination, the present study examined the effects of σR antagonist and DAT inhibitor combinations in male rats discriminating cocaine (10 mg/kg, i.p.) from saline injections. The DAT inhibitors alone [(-)-2-β-carbomethoxy-3-β-(4-fluorophenyl)tropane 1,5-naphthalenedisulfonate monohydrate (WIN 35,428) and methylphenidate] at low (0.1-mg/kg) doses that were minimally active failed to shift the dose-effect function for discriminative-stimulus effects of cocaine to the left more than 2-fold. At 0.32 mg/kg the DAT inhibitors alone shifted the cocaine dose-effect function leftward 24- or 6.6-fold, respectively. The σR antagonists (BD1008, BD1047, and BD1063) failed to fully substitute for cocaine, although BD1008 and BD1047 substituted partially. At 10 mg/kg, BD1008, BD1047, or BD1063 alone shifted the cocaine dose-effect function leftward less than 6.0-fold. In combination with 0.1 mg/kg WIN 35,428, the 10 mg/kg doses of σR antagonists shifted the cocaine dose-effect function from 12.3- to 36.7-fold leftward, and with 0.32 mg/kg WIN 35,428 from 14.3- to 440-fold leftward. In combination with 0.1 mg/kg methylphenidate, those σR antagonist doses shifted the cocaine dose-effect function from 5.5- to 55.0-fold leftward, and with 0.32 mg/kg methylphenidate from 10.5- to 48.1-fold leftward. The present results suggest that dual DAT/σR inhibition produces agonist-like subjective effects that may promote decreases in self-administration obtained in previous studies. SIGNIFICANCE STATEMENT: There is currently no approved medication for treating stimulant abuse, although dopamine uptake inhibitors in combination with sigma receptor (σR) antagonists decrease cocaine self-administration in laboratory animals. The present study assessed how this combination alters the discriminative-stimulus effects of cocaine in male rats. Results suggest that concurrent dopamine uptake inhibition and σR antagonism together may promote decreases in self-administration, possibly by mimicking the subjective effects extant when subjects cease continued cocaine self-administration.
Collapse
Affiliation(s)
- Takato Hiranita
- Department of Pharmacology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas (T.H.) and Psychobiology Section, Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland (S.-M.L., J.L.K.)
| | - Su-Min Li
- Department of Pharmacology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas (T.H.) and Psychobiology Section, Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland (S.-M.L., J.L.K.)
| | - Jonathan L Katz
- Department of Pharmacology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas (T.H.) and Psychobiology Section, Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland (S.-M.L., J.L.K.)
| |
Collapse
|
2
|
Obeng S, Crowley ML, Mottinelli M, León F, Zuarth Gonzalez JD, Chen Y, Gamez-Jimenez LR, Restrepo LF, Ho NP, Patel A, Martins Rocha J, Alvarez MA, Thadisetti AM, Park CR, Pallares VLC, Milner MJ, Canal CE, Hampson AJ, McCurdy CR, McMahon LR, Wilkerson JL, Hiranita T. The Mitragyna speciosa (kratom) alkaloid mitragynine: Analysis of adrenergic α 2 receptor activity in vitro and in vivo. Eur J Pharmacol 2024; 980:176863. [PMID: 39068978 DOI: 10.1016/j.ejphar.2024.176863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
Mitragynine, an alkaloid present in the leaves of Mitragyna speciosa (kratom), has a complex pharmacology that includes low efficacy agonism at μ-opioid receptors (MORs). This study examined the activity of mitragynine at adrenergic α2 receptors (Aα2Rs) in vitro and in vivo. Mitragynine displaced a radiolabeled Aα2R antagonist ([3H]RX821002) from human Aα2ARs in vitro with lower affinity (Ki = 1260 nM) than the agonists (-)-epinephrine (Ki = 263 nM) or lofexidine (Ki = 7.42 nM). Mitragynine did not significantly stimulate [35S]GTPγS binding at Aα2ARs in vitro, but in rats trained to discriminate 32 mg/kg mitragynine from vehicle (intraperitoneally administered; i.p.), mitragynine exerted an Aα2R agonist-like effect. Both α2R antagonists (atipamezole and yohimbine) and MOR antagonists (naloxone and naltrexone) produced rightward shifts in mitragynine discrimination dose-effect function and Aα2R agonists lofexidine and clonidine produced leftward shifts. In the mitragynine trained rats, Aα2R agonists also produced leftward shifts in discrimination dose-effect functions for morphine and fentanyl. In a separate rat cohort trained to discriminate 3.2 mg/kg i.p. morphine from vehicle, naltrexone produced a rightward shift, but neither an Aα2R agonist or antagonist affected morphine discrimination. In a hypothermia assay, both lofexidine and clonidine produced marked effects antagonized by yohimbine. Mitragynine did not produce hypothermia. Together, these data demonstrate that mitragynine acts in vivo like an Aα2R agonist, although its failure to induce hypothermia or stimulate [35S]GTPγS binding in vitro, suggests that mitragynine maybe a low efficacy Aα2R agonist.
Collapse
Affiliation(s)
- Samuel Obeng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA; Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Morgan L Crowley
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Marco Mottinelli
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Francisco León
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Julio D Zuarth Gonzalez
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Yiming Chen
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Atlanta, GA, 30341, USA
| | - Lea R Gamez-Jimenez
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Luis F Restrepo
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Nicholas P Ho
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Avi Patel
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Joelma Martins Rocha
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Manuel A Alvarez
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Amsha M Thadisetti
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Chai R Park
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Victoria L C Pallares
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Megan J Milner
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Clinton E Canal
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Atlanta, GA, 30341, USA
| | - Aidan J Hampson
- Division of Therapeutics and Medical Consequences, National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA; Department of Pharmaceutics, University of Florida, Gainesville, FL 32610, USA; Translational Drug Development Core, Clinical and Translational Sciences Institute, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Lance R McMahon
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Jenny L Wilkerson
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA.
| | - Takato Hiranita
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
3
|
Yates JR. Pharmacological Treatments for Methamphetamine Use Disorder: Current Status and Future Targets. Subst Abuse Rehabil 2024; 15:125-161. [PMID: 39228432 PMCID: PMC11370775 DOI: 10.2147/sar.s431273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024] Open
Abstract
The illicit use of the psychostimulant methamphetamine (METH) is a major concern, with overdose deaths increasing substantially since the mid-2010s. One challenge to treating METH use disorder (MUD), as with other psychostimulant use disorders, is that there are no available pharmacotherapies that can reduce cravings and help individuals achieve abstinence. The purpose of the current review is to discuss the molecular targets that have been tested in assays measuring the physiological, the cognitive, and the reinforcing effects of METH in both animals and humans. Several drugs show promise as potential pharmacotherapies for MUD when tested in animals, but fail to produce long-term changes in METH use in dependent individuals (eg, modafinil, antipsychotic medications, baclofen). However, these drugs, plus medications like atomoxetine and varenicline, may be better served as treatments to ameliorate the psychotomimetic effects of METH or to reverse METH-induced cognitive deficits. Preclinical studies show that vesicular monoamine transporter 2 inhibitors, metabotropic glutamate receptor ligands, and trace amine-associated receptor agonists are efficacious in attenuating the reinforcing effects of METH; however, clinical studies are needed to determine if these drugs effectively treat MUD. In addition to screening these compounds in individuals with MUD, potential future directions include increased emphasis on sex differences in preclinical studies and utilization of pharmacogenetic approaches to determine if genetic variances are predictive of treatment outcomes. These future directions can help lead to better interventions for treating MUD.
Collapse
Affiliation(s)
- Justin R Yates
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY, USA
| |
Collapse
|
4
|
Strumberger CD, D'Epagnier EJ, Nguyen KH, Rogers JD, Meyer MP, Malhotra Y, Hinman JE, Jansen EL, Minervini V. Antinociceptive and adverse effects of morphine:ketamine mixtures in rats. Behav Pharmacol 2024; 35:122-131. [PMID: 38451024 DOI: 10.1097/fbp.0000000000000761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Prescription opioids are the gold standard for treating moderate to severe pain despite their well-documented adverse effects. Of all prescription medications, opioids are abused most widely, and fatal overdoses have reached epidemic levels. One strategy for improving the margin of safety of opioids is combining them with non-opioid drugs to decrease the opioid dose needed for pain relief, thereby reducing adverse effects that occur with larger doses. The N-methyl-D-aspartate receptor antagonist ketamine has been used safely as an analgesic but only under a very limited range of conditions. The current studies characterized the antinociceptive, behavioral suppressant, and gastrointestinal effects of morphine and ketamine alone and in mixtures to determine their interaction in 24 adult male Sprague-Dawley rats (n = 8 per assay). Given alone, both morphine and ketamine produced antinociception, decreased responding for food, and reduced gastrointestinal transit (i.e. produced constipation). The effects of morphine:ketamine mixtures generally were additive, except for the antinociceptive effects of 1:1 mixtures for which the difference in slope (i.e. non-parallel shift) between the observed and predicted effects suggested synergy at smaller doses and additivity at larger doses. The potency of morphine to produce constipation was not enhanced by administration of morphine:ketamine mixtures with antinociceptive effects. The nature of the interaction between morphine and ketamine for adverse effects such as dependence, withdrawal, abuse, or respiratory depression remains unknown but also might be related to the ratio of each drug in mixtures. It will be important to identify conditions that produce the largest potential therapeutic window in humans.
Collapse
Affiliation(s)
- Conor D Strumberger
- Department of Psychological Science, Creighton University, Omaha, Nebraska, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Knowles LG, Armanious AJ, Peng Y, Welsh WJ, James MH. Recent advances in drug discovery efforts targeting the sigma 1 receptor system: Implications for novel medications designed to reduce excessive drug and food seeking. ADDICTION NEUROSCIENCE 2023; 8:100126. [PMID: 37753198 PMCID: PMC10519676 DOI: 10.1016/j.addicn.2023.100126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Psychiatric disorders characterized by uncontrolled reward seeking, such as substance use disorders (SUDs), alcohol use disorder (AUD) and some eating disorders, impose a significant burden on individuals and society. Despite their high prevalence and substantial morbidity and mortality rates, treatment options for these disorders remain limited. Over the past two decades, there has been a gradual accumulation of evidence pointing to the sigma-1 receptor (S1R) system as a promising target for therapeutic interventions designed to treat these disorders. S1R is a chaperone protein that resides in the endoplasmic reticulum, but under certain conditions translocates to the plasma membrane. In the brain, S1Rs are expressed in several regions important for reward, and following translocation, they physically associate with several reward-related GPCRs, including dopamine receptors 1 and 2 (D1R and D2R). Psychostimulants, alcohol, as well as palatable foods, all alter expression of S1R in regions important for motivated behavior, and S1R antagonists generally decrease behavioral responses to these rewards. Recent advances in structural modeling have permitted the development of highly-selective S1R antagonists with favorable pharmacokinetic profiles, thus providing a therapeutic avenue for S1R-based medications. Here, we provide an up-to-date overview of work linking S1R with motivated behavior for drugs of abuse and food, as well as evidence supporting the clinical utility of S1R antagonists to reduce their excessive consumption. We also highlight potential challenges associated with targeting the S1R system, including the need for a more comprehensive understanding of the underlying neurobiology and careful consideration of the pharmacological properties of S1R-based drugs.
Collapse
Affiliation(s)
- Liam G. Knowles
- Harpur School of Arts and Sciences, Binghamton University, Vestal, NY, USA
| | - Abanoub J. Armanious
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Youyi Peng
- Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - William J. Welsh
- Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA
| | - Morgan H. James
- Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University and Rutgers Biomedical Health Sciences, Piscataway, NJ, USA
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| |
Collapse
|
6
|
Jensen KL, Jensen SB, Madsen KL. A mechanistic overview of approaches for the treatment of psychostimulant dependence. Front Pharmacol 2022; 13:854176. [PMID: 36160447 PMCID: PMC9493975 DOI: 10.3389/fphar.2022.854176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Psychostimulant use disorder is a major health issue around the world with enormous individual, family-related and societal consequences, yet there are no effective pharmacological treatments available. In this review, a target-based overview of pharmacological treatments toward psychostimulant addiction will be presented. We will go through therapeutic approaches targeting different aspects of psychostimulant addiction with focus on three major areas; 1) drugs targeting signalling, and metabolism of the dopamine system, 2) drugs targeting either AMPA receptors or metabotropic glutamate receptors of the glutamate system and 3) drugs targeting the severe side-effects of quitting long-term psychostimulant use. For each of these major modes of intervention, findings from pre-clinical studies in rodents to clinical trials in humans will be listed, and future perspectives of the different treatment strategies as well as their potential side-effects will be discussed. Pharmaceuticals modulating the dopamine system, such as antipsychotics, DAT-inhibitors, and disulfiram, have shown some promising results. Cognitive enhancers have been found to increase aspects of behavioural control, and drugs targeting the glutamate system such as modulators of metabotropic glutamate receptors and AMPA receptors have provided interesting changes in relapse behaviour. Furthermore, CRF-antagonists directed toward alleviating the symptoms of the withdrawal stage have been examined with interesting resulting changes in behaviour. There are promising results investigating therapeutics for psychostimulant addiction, but further preclinical work and additional human studies with a more stratified patient selection are needed to prove sufficient evidence of efficacy and tolerability.
Collapse
|
7
|
Mathieson E, Irving C, Koberna S, Nicholson M, Otto MW, Kantak KM. Role of preexisting inhibitory control deficits vs. drug use history in mediating insensitivity to aversive consequences in a rat model of polysubstance use. Psychopharmacology (Berl) 2022; 239:2377-2394. [PMID: 35391547 PMCID: PMC8989405 DOI: 10.1007/s00213-022-06134-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 03/30/2022] [Indexed: 10/26/2022]
Abstract
RATIONALE The nature and predictors of insensitivity to aversive consequences of heroin + cocaine polysubstance use are not well characterized. OBJECTIVES Translational methods incorporating a tightly controlled animal model of drug self-administration and measures of inhibitory control and avoidance behavior might be helpful for clarifying this issue. METHODS The key approach for distinguishing potential contributions of pre-existing inhibitory control deficits vs. drug use history in meditating insensitivity to aversive consequences was comparison of two rat strains: Wistar (WIS/Crl), an outbred strain, and the spontaneously hypertensive rat (SHR/NCrl), an inbred strain shown previously to exhibit heightened cocaine and heroin self-administration and poor inhibitory control relative to WIS/Crl. RESULTS In separate tasks, SHR/NCrl displayed greater impulsive action and compulsive-like behavior than WIS/Crl prior to drug exposure. Under two different schedules of drug delivery, SHR/NCrl self-administered more cocaine than WIS/Crl, but self-administered a similar amount of heroin + cocaine as WIS/Crl. When half the session cycles were punished by random foot shock, SHR/NCrl initially were less sensitive to punishment than WIS/Crl when self-administering cocaine, but were similarly insensitive to punishment when self-administering heroin + cocaine. Based on correlation analyses, only trait impulsivity predicted avoidance capacity in rats self-administering cocaine and receiving yoked-saline. In contrast, only amount of drug use predicted avoidance capacity in rats self-administering heroin + cocaine. Additionally, baseline drug seeking and taking predicted punishment insensitivity in rats self-administering cocaine or heroin + cocaine. CONCLUSIONS Based on the findings revealed in this animal model, human laboratory research concerning the nature and predictors of insensitivity to aversive consequences in heroin and cocaine polysubstance vs. monosubstance users is warranted.
Collapse
Affiliation(s)
- Elon Mathieson
- Department of Psychological and Brain Sciences, Boston University, 64 Cummington Mall, Boston, MA, 02215, USA
| | - Carolyn Irving
- Department of Psychological and Brain Sciences, Boston University, 64 Cummington Mall, Boston, MA, 02215, USA
| | - Sarah Koberna
- Department of Psychological and Brain Sciences, Boston University, 64 Cummington Mall, Boston, MA, 02215, USA
| | - Megan Nicholson
- Department of Psychological and Brain Sciences, Boston University, 64 Cummington Mall, Boston, MA, 02215, USA
| | - Michael W Otto
- Department of Psychological and Brain Sciences, Boston University, 64 Cummington Mall, Boston, MA, 02215, USA
| | - Kathleen M Kantak
- Department of Psychological and Brain Sciences, Boston University, 64 Cummington Mall, Boston, MA, 02215, USA.
- Center for Systems Neuroscience, Boston University, Boston, MA, USA.
| |
Collapse
|
8
|
Interactions between opioids and stimulants: Behavioral pharmacology of abuse-related effects. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 93:1-33. [PMID: 35341563 DOI: 10.1016/bs.apha.2021.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Opioid abuse continues to be a significant public health challenge, with rates of opioid-related overdose deaths increasing continuously over the last two decades. There also has been a sharp increase in overdose deaths involving stimulant drugs, primarily cocaine and methamphetamine. Recent estimates indicate a high prevalence of co-use of opioids and stimulants, which is a particularly complex problem. Behavioral pharmacology research over the last few decades has characterized interactions between opioids and stimulants as well as evaluated potential treatments. This chapter describes interactions between opioids and stimulants, with a focus on pre-clinical studies of abuse-related behavioral effects using self-administration, reinstatement, drug discrimination, place conditioning, and intracranial self-stimulation paradigms in laboratory animals. In general, the literature provides substantial evidence of mutual enhancement between opioids and stimulants for abuse-related effects, although such results are not ubiquitous. Enhanced abuse-related effects could manifest in many ways including engaging in drug seeking and taking behaviors with greater persistence, effort, and motivation and/or increased likelihood of relapse. Moreover, studies on opioid/stimulant combinations set the stage for evaluating potential treatments for polysubstance use. Behavioral pharmacology research has proven invaluable for elucidating these relationships using rigorous experimental designs and quantitative analyses of pharmacological and behavioral data.
Collapse
|
9
|
Physiological dependence to mitragynine indicated by a rapid cross-dependence procedure with heroin-dependent mice. Psychopharmacology (Berl) 2022; 239:897-908. [PMID: 35107609 DOI: 10.1007/s00213-022-06080-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/27/2022] [Indexed: 10/19/2022]
Abstract
The potential of mitragynine to produce physiological dependence (withdrawal) was assessed using a rapid assessment procedure with male ICR mice exposed to heroin-admixed food followed by naloxone (subcutaneously, s.c.) precipitation of withdrawal. Initial studies indicated that 3 days of exposure to 3.0 mg/g of heroin-admixed food followed by naloxone (0.6 mg/kg) reliably precipitated withdrawal jumping and weight loss. Lower concentrations of heroin-admixed food and lower doses of naloxone produced fewer withdrawal signs. A longer exposure to heroin-admixed food did not produce significantly greater amounts of jumping or weight loss. Further, these withdrawal signs were dose-dependently reversed by s.c. administration of heroin immediately following naloxone administration. Mitragynine (s.c.) also dose-dependently suppressed naloxone-precipitated withdrawal signs. Additionally, both jumping and weight loss were suppressed over a comparable range of mitragynine doses when administered by gavage with a noticeably, but not significantly, higher potency than with s.c. administration. The ED50 values for mitragynine for the suppression of withdrawal by any route (354-911 μmol/kg) were greater than the minimally effective dose that decreased locomotor activity (251 μmol/kg) and from 40- to 104-fold greater than those for heroin. The results suggest inherent opioid dependence liability of mitragynine. The in vivo potency relations between mitragynine and heroin are consistent with a conclusion of dependence-producing effects, indicated by the suppression of withdrawal, comparable to standard opioid μ-receptor agonists, differing primarily in terms of potency. The present paper provides a method for the rapid assessment of physiological dependence liability applicable to other kratom plant constituents or any potential opioid dependence-producing agents.
Collapse
|
10
|
Hiranita T, Obeng S, Sharma A, Wilkerson JL, McCurdy CR, McMahon LR. In vitro and in vivo pharmacology of kratom. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 93:35-76. [PMID: 35341571 DOI: 10.1016/bs.apha.2021.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Kratom products have been historically and anecdotally used in south Asian countries for centuries to manage pain and opioid withdrawal. The use of kratom products has dramatically increased in the United States. More than 45 kratom alkaloids have been isolated, yet the overall pharmacology of the individual alkaloids is still not well characterized. The purpose of this chapter is to summarize in vitro and in vivo opioid activities of the primary kratom alkaloid mitragynine and its more potent metabolite 7-hydroxymitragynine. Following are experimental procedures described to characterize opioid receptor activity; receptor binding and functional assays, antinociceptive assays, operant conditioning assays, and respiratory plethysmography. The capacity of kratom alkaloids to confer tolerance and physical dependence as well as their pharmacokinetic properties are also summarized. The data reviewed here suggest that kratom products and mitragynine possess low efficacy agonist activity at the mu-opioid receptor in vivo. In addition, kratom products and mitragynine have been demonstrated to antagonize the effects of high efficacy mu-opioid agonists. The data further suggest that 7-hydroxymitragynine formed in vivo by metabolism of mitragynine may be minimally involved in the overall behavioral profile of mitragynine and kratom, whereas 7-hydroxymitragynine itself, at sufficiently high doses administered exogenously, shares many of the same abuse- and dependence-related behavioral effects associated with traditional opioid agonists. The apparent low efficacy of kratom products and mitragynine at mu-opioid receptors supports the development of these ligands as effective and potentially safe medications for opioid use disorder.
Collapse
Affiliation(s)
- Takato Hiranita
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Samuel Obeng
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States; Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Abhisheak Sharma
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, United States; Translational Drug Development Core, Clinical and Translational Sciences Institute, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Jenny L Wilkerson
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, United States; Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, United States; Translational Drug Development Core, Clinical and Translational Sciences Institute, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Lance R McMahon
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
11
|
Banks ML. Environmental influence on the preclinical evaluation of substance use disorder therapeutics. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 93:219-242. [PMID: 35341567 DOI: 10.1016/bs.apha.2021.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Substance use disorders (SUD) develop as a result of complex interactions between the environment, the subject, and the drug of abuse. Preclinical basic research investigating each of these tripartite components of SUD individually has resulted in advancements in our fundamental knowledge regarding the progression from drug abuse to SUD and severe drug addiction and the underlying behavioral and neurobiological mechanisms. How these complex interactions between the environment, the subject, and the drug of abuse impact the effectiveness of candidate or clinically used medications for SUD has not been as extensively investigated. The focus of this chapter will address the current state of our knowledge how these environmental, subject, and pharmacological variables have been shown to impact candidate or clinical SUD medication evaluation in preclinical research using drug self-administration procedures as the primary dependent measure. The results discussed in this chapter highlight the importance of considering environmental variables such as the schedule of reinforcement, concurrent availability of alternative nondrug reinforcers, and experimental housing conditions in the context of SUD therapeutic evaluation. The thesis of this chapter is that improved understanding of environmental variables in the context of SUD research will facilitate the utility of preclinical drug self-administration studies in the evaluation and development of candidate SUD therapeutics.
Collapse
Affiliation(s)
- Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.
| |
Collapse
|
12
|
Obeng S, Wilkerson JL, León F, Reeves ME, Restrepo LF, Gamez-Jimenez LR, Patel A, Pennington AE, Taylor VA, Ho NP, Braun T, Fortner JD, Crowley ML, Williamson MR, Pallares VLC, Mottinelli M, Lopera-Londoño C, McCurdy CR, McMahon LR, Hiranita T. Pharmacological Comparison of Mitragynine and 7-Hydroxymitragynine: In Vitro Affinity and Efficacy for μ-Opioid Receptor and Opioid-Like Behavioral Effects in Rats. J Pharmacol Exp Ther 2020; 376:410-427. [PMID: 33384303 DOI: 10.1124/jpet.120.000189] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/28/2020] [Indexed: 01/30/2023] Open
Abstract
Relationships between µ-opioid receptor (MOR) efficacy and effects of mitragynine and 7-hydroxymitragynine are not fully established. We assessed in vitro binding affinity and efficacy and discriminative stimulus effects together with antinociception in rats. The binding affinities of mitragynine and 7-hydroxymitragynine at MOR (Ki values 77.9 and 709 nM, respectively) were higher than their binding affinities at κ-opioid receptor (KOR) or δ-opioid receptor (DOR). [35S]guanosine 5'-O-[γ-thio]triphosphate stimulation at MOR demonstrated that mitragynine was an antagonist, whereas 7-hydroxymitragynine was a partial agonist (Emax = 41.3%). In separate groups of rats discriminating either morphine (3.2 mg/kg) or mitragynine (32 mg/kg), mitragynine produced a maximum of 72.3% morphine-lever responding, and morphine produced a maximum of 65.4% mitragynine-lever responding. Other MOR agonists produced high percentages of drug-lever responding in the morphine and mitragynine discrimination assays: 7-hydroxymitragynine (99.7% and 98.1%, respectively), fentanyl (99.7% and 80.1%, respectively), buprenorphine (99.8% and 79.4%, respectively), and nalbuphine (99.4% and 98.3%, respectively). In the morphine and mitragynine discrimination assays, the KOR agonist U69,593 produced maximums of 72.3% and 22.3%, respectively, and the DOR agonist SNC 80 produced maximums of 34.3% and 23.0%, respectively. 7-Hydroxymitragynine produced antinociception; mitragynine did not. Naltrexone antagonized all of the effects of morphine and 7-hydroxymitragynine; naltrexone antagonized the discriminative stimulus effects of mitragynine but not its rate-decreasing effects. Mitragynine increased the potency of the morphine discrimination yet decreased morphine antinociception. Here we illustrate striking differences in MOR efficacy, with mitragynine having less than 7-hydroxymitragynine. SIGNIFICANCE STATEMENT: At human µ-opioid receptor (MOR) in vitro, mitragynine has low affinity and is an antagonist, whereas 7-hydroxymitragynine has 9-fold higher affinity than mitragynine and is an MOR partial agonist. In rats, intraperitoneal mitragynine exhibits a complex pharmacology including MOR agonism; 7-hydroxymitragynine has higher MOR potency and efficacy than mitragynine. These results are consistent with 7-hydroxymitragynine being a highly selective MOR agonist and with mitragynine having a complex pharmacology that combines low efficacy MOR agonism with activity at nonopioid receptors.
Collapse
Affiliation(s)
- Samuel Obeng
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Jenny L Wilkerson
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Francisco León
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Morgan E Reeves
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Luis F Restrepo
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Lea R Gamez-Jimenez
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Avi Patel
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Anna E Pennington
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Victoria A Taylor
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Nicholas P Ho
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Tobias Braun
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - John D Fortner
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Morgan L Crowley
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Morgan R Williamson
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Victoria L C Pallares
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Marco Mottinelli
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Carolina Lopera-Londoño
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Christopher R McCurdy
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Lance R McMahon
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| | - Takato Hiranita
- Departments of Pharmacodynamics (S.O., J.L.W., M.E.R., L.F.R., L.R.G.-J., A.P., A.E.P., V.A.T., N.P.H., T.B., M.R.W., V.L.C.P., L.R.M., T.H.) and Medicinal Chemistry (S.O., F.L., J.D.F., M.L.C., M.M., C.L.-L., C.R.M.), and Translational Drug Development Core, Clinical and Translational Sciences Institutes (C.R.M.), College of Pharmacy, University of Florida, Gainesville, Florida
| |
Collapse
|
13
|
Soto PL, Hiranita T. Effects of benztropine analogs on delay discounting in rats. Psychopharmacology (Berl) 2020; 237:3783-3794. [PMID: 32964243 PMCID: PMC7686108 DOI: 10.1007/s00213-020-05655-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 08/31/2020] [Indexed: 10/23/2022]
Abstract
RATIONALE Methylphenidate and d-amphetamine, medications used for treatment of attention deficit hyperactivity disorder (ADHD), are used recreationally and self-administered by laboratory animals. Benztropine (BZT) analogs, like those medications, increase synaptic dopamine levels but are less effective in maintaining self-administration, suggesting clinical utility with less abuse liability. OBJECTIVES The current study was designed to evaluate potential therapeutic effects of BZT analogs related to ADHD. METHODS Rats responded under a delay-discounting procedure in which responses on one lever produced immediate delivery of a single food pellet and alternative responses produced four food pellets either immediately or with various temporal delays, with those delays arranged in ascending or random orders in different groups of rats. Selection of the smaller more immediate reinforcer has been suggested as an aspect of "impulsivity," a trait with suggested involvement in ADHD. Other rats were studied under fixed-interval (FI) 300-s schedules to assess drug effects on behavior under temporal control. RESULTS d-Amphetamine, methylphenidate, and the BZT analog AHN 1-055, but not AHN 2-005 or JHW 007, increased selection of the large, delayed reinforcer with either arrangement of delays. All drugs changed the temporal distribution of responses within the FI from one with responses concentrated at the end to a more uniform distribution. Changes in the temporal distribution of FI responding occurred with drugs that did not affect discounting suggesting that discounting does not arise directly from the same temporal control processes controlling FI responding. CONCLUSIONS AHN 1-055 may be of clinical utility in the treatment of ADHD.
Collapse
Affiliation(s)
- Paul L Soto
- Department of Psychology, Louisiana State University, Baton Rouge, LA, 70803, USA.
| | - Takato Hiranita
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| |
Collapse
|
14
|
Abdullah CS, Aishwarya R, Alam S, Morshed M, Remex NS, Nitu S, Kolluru GK, Traylor J, Miriyala S, Panchatcharam M, Hartman B, King J, Bhuiyan MAN, Chandran S, Woolard MD, Yu X, Goeders NE, Dominic P, Arnold CL, Stokes K, Kevil CG, Orr AW, Bhuiyan MS. Methamphetamine induces cardiomyopathy by Sigmar1 inhibition-dependent impairment of mitochondrial dynamics and function. Commun Biol 2020; 3:682. [PMID: 33203971 PMCID: PMC7673131 DOI: 10.1038/s42003-020-01408-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Methamphetamine-associated cardiomyopathy is the leading cause of death linked with illicit drug use. Here we show that Sigmar1 is a therapeutic target for methamphetamine-associated cardiomyopathy and defined the molecular mechanisms using autopsy samples of human hearts, and a mouse model of "binge and crash" methamphetamine administration. Sigmar1 expression is significantly decreased in the hearts of human methamphetamine users and those of "binge and crash" methamphetamine-treated mice. The hearts of methamphetamine users also show signs of cardiomyopathy, including cellular injury, fibrosis, and enlargement of the heart. In addition, mice expose to "binge and crash" methamphetamine develop cardiac hypertrophy, fibrotic remodeling, and mitochondrial dysfunction leading to contractile dysfunction. Methamphetamine treatment inhibits Sigmar1, resulting in inactivation of the cAMP response element-binding protein (CREB), decreased expression of mitochondrial fission 1 protein (FIS1), and ultimately alteration of mitochondrial dynamics and function. Therefore, Sigmar1 is a viable therapeutic agent for protection against methamphetamine-associated cardiomyopathy.
Collapse
Affiliation(s)
- Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Richa Aishwarya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Shafiul Alam
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Sadia Nitu
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Gopi K Kolluru
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - James Traylor
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Sumitra Miriyala
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Manikandan Panchatcharam
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Brandon Hartman
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Judy King
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | | | - Sunitha Chandran
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Matthew D Woolard
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Xiuping Yu
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Nicholas E Goeders
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Paari Dominic
- Department of Medicine, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Connie L Arnold
- Department of Medicine, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Karen Stokes
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Christopher G Kevil
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - A Wayne Orr
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA.
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA.
| |
Collapse
|
15
|
Behnood-Rod A, Chellian R, Wilson R, Hiranita T, Sharma A, Leon F, McCurdy CR, McMahon LR, Bruijnzeel AW. Evaluation of the rewarding effects of mitragynine and 7-hydroxymitragynine in an intracranial self-stimulation procedure in male and female rats. Drug Alcohol Depend 2020; 215:108235. [PMID: 32889450 PMCID: PMC7542979 DOI: 10.1016/j.drugalcdep.2020.108235] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/28/2020] [Accepted: 08/11/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Kratom (Mitragyna speciosa Korth.) has been used in Southeast Asia for hundreds of years to increase energy, for relaxation, and to diminish opioid withdrawal. Kratom use has recently spread to Western countries. Kratom could potentially be used for the treatment of opioid withdrawal and pain, but more insight is needed into its abuse potential. Therefore, we investigated the rewarding properties of the primary kratom alkaloid mitragynine and its active metabolite 7-hydroxymitragynine, and morphine as a reference drug in male and female rats. These compounds have agonist activity at mu-opioid receptors. METHODS The compounds were tested in an intracranial self-stimulation (ICSS) procedure, which allows for the evaluation of the rewarding/aversive and sedative effects of drugs. Rewarding doses of drugs decrease the brain reward thresholds, and aversive drug doses have the opposite effect. RESULTS Mitragynine, 7-hydroxymitragynine, and morphine affected the brain reward thresholds. A high dose of 7-hydroxymitragynine (3.2 mg/kg) increased the brain reward thresholds, whereas an intermediate dose of morphine (10 mg/kg) decreased the reward thresholds. 7-Hydroxymitragynine and morphine affected the response latencies. Five mg/kg of morphine increased response latencies. 7-Hydroxymitragynine tended to increase the response latencies, but the post hoc analyses did not reveal a significant effect. There were no sex differences in the effects of mitragynine, 7-hydroxymitragynine, and morphine on the reward thresholds and the response latencies. CONCLUSIONS These initial findings indicate that mitragynine and 7-hydroxymitragynine are not rewarding in the ICSS procedure. The present results suggest that these kratom alkaloids do not have abuse potential.
Collapse
Affiliation(s)
- Azin Behnood-Rod
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | | | - Ryann Wilson
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Takato Hiranita
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Abhisheak Sharma
- Translational Drug Development Core, Clinical and Translational Sciences Institute, University of Florida, Gainesville, FL, USA,Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Francisco Leon
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Christopher R. McCurdy
- Translational Drug Development Core, Clinical and Translational Sciences Institute, University of Florida, Gainesville, FL, USA,Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Lance R. McMahon
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | | |
Collapse
|
16
|
Modafinil potentiates cocaine self-administration by a dopamine-independent mechanism: possible involvement of gap junctions. Neuropsychopharmacology 2020; 45:1518-1526. [PMID: 32340023 PMCID: PMC7360549 DOI: 10.1038/s41386-020-0680-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/15/2020] [Accepted: 04/09/2020] [Indexed: 12/21/2022]
Abstract
Modafinil and methylphenidate are medications that inhibit the neuronal reuptake of dopamine, a mechanism shared with cocaine. Their use as "smart drugs" by healthy subjects poses health concerns and requires investigation. We show that methylphenidate, but not modafinil, maintained intravenous self-administration in Sprague-Dawley rats similar to cocaine. Both modafinil and methylphenidate pretreatments potentiated cocaine self-administration. Cocaine, at self-administered doses, stimulated mesolimbic dopamine levels. This effect was potentiated by methylphenidate, but not by modafinil pretreatments, indicating dopamine-dependent actions for methylphenidate, but not modafinil. Modafinil is known to facilitate electrotonic neuronal coupling by actions on gap junctions. Carbenoxolone, a gap junction inhibitor, antagonized modafinil, but not methylphenidate potentiation of cocaine self-administration. Our results indicate that modafinil shares mechanisms with cocaine and methylphenidate but has a unique pharmacological profile that includes facilitation of electrotonic coupling and lower abuse liability, which may be exploited in future therapeutic drug design for cocaine use disorder.
Collapse
|
17
|
The discriminative stimulus effects of epibatidine in C57BL/6J mice. Behav Pharmacol 2020; 31:565-573. [PMID: 32209809 DOI: 10.1097/fbp.0000000000000555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The α4β2* nicotinic acetylcholine receptor (nAChR) subtypes are targeted for the development of smoking cessation aids, and the use of drug discrimination in mice provides a robust screening tool for the identification of drugs acting through nAChRs. Here, we established that the α4β2* nAChR agonist epibatidine can function as a discriminative stimulus in mice. Male C57BL/6J mice discriminated epibatidine (0.0032 mg/kg, subcutaneously) and were tested with agonists varying in selectivity and efficacy for α4β2* nAChRs. The discriminative stimulus effects of epibatidine were characterized with the nonselective, noncompetitive nicotinic antagonist mecamylamine, with the selective β2-substype-containing nAChR antagonist dihydro-β-erythroidine hydrobromide (DHβE), and the α7 antagonist methyllycaconitine (MLA). Nicotine (0.32-1.0 mg/kg, subcutaneously), the partial nAChR agonist cytisine (1.0-5.6 mg/kg, subcutaneously), and the α7 nAChR agonist N-[(3R)-1-azabicyclo[2.2.2]oct-3-yl]-4-chlorobenzamide (10-56 mg/kg, intraperitoneally) produced no more than 33% epibatidine-appropriate responding. The partial α4β2* nAChR agonists varenicline and 2'-fluoro-3'-(4-nitro-phenyl)deschloroepibatidine produced 61 and 69% epibatidine-appropriate responding, respectively. DHβE and mecamylamine, but not MLA, significantly antagonized the discriminative stimulus effects of epibatidine. These results show that epibatidine may be trained as a discriminative stimulus in mice and has utility in elucidating the in-vivo pharmacology of α4β2* nAChR ligands.
Collapse
|
18
|
Slack RD, Ku T, Cao J, Giancola J, Bonifazi A, Loland CJ, Gadiano A, Lam J, Rais R, Slusher BS, Coggiano M, Tanda G, Newman AH. Structure-Activity Relationships for a Series of (Bis(4-fluorophenyl)methyl)sulfinyl Alkyl Alicyclic Amines at the Dopamine Transporter: Functionalizing the Terminal Nitrogen Affects Affinity, Selectivity, and Metabolic Stability. J Med Chem 2020; 63:2343-2357. [PMID: 31661268 PMCID: PMC9617638 DOI: 10.1021/acs.jmedchem.9b01188] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Atypical dopamine transporter (DAT) inhibitors have shown therapeutic potential in preclinical models of psychostimulant abuse. In rats, 1-(4-(2-((bis(4-fluorophenyl)methyl)sulfinyl)ethyl)-piperazin-1-yl)-propan-2-ol (3b) was effective in reducing the reinforcing effects of both cocaine and methamphetamine but did not exhibit psychostimulant behaviors itself. While further development of 3b is ongoing, diastereomeric separation, as well as improvements in potency and pharmacokinetics were desirable for discovering pipeline drug candidates. Thus, a series of bis(4-fluorophenyl)methyl)sulfinyl)alkyl alicyclic amines, where the piperazine-2-propanol scaffold was modified, were designed, synthesized, and evaluated for binding affinities at DAT, as well as the serotonin transporter and σ1 receptors. Within the series, 14a showed improved DAT affinity (Ki = 23 nM) over 3b (Ki = 230 nM), moderate metabolic stability in human liver microsomes, and a hERG/DAT affinity ratio = 28. While 14a increased locomotor activity relative to vehicle, it was significantly lower than activity produced by cocaine. These results support further investigation of 14a as a potential treatment for psychostimulant use disorders.
Collapse
Affiliation(s)
- Rachel D. Slack
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Therese Ku
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - JoLynn Giancola
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Alessandro Bonifazi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Claus J. Loland
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Alexandra Gadiano
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, United States
| | - Jenny Lam
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, United States
| | - Rana Rais
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, United States
| | - Barbara S. Slusher
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, United States
| | - Mark Coggiano
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Gianluigi Tanda
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| |
Collapse
|
19
|
Jordan CJ, Cao J, Newman AH, Xi ZX. Progress in agonist therapy for substance use disorders: Lessons learned from methadone and buprenorphine. Neuropharmacology 2019; 158:107609. [PMID: 31009632 PMCID: PMC6745247 DOI: 10.1016/j.neuropharm.2019.04.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/25/2019] [Accepted: 04/12/2019] [Indexed: 12/12/2022]
Abstract
Substance use disorders (SUD) are serious public health problems worldwide. Although significant progress has been made in understanding the neurobiology of drug reward and the transition to addiction, effective pharmacotherapies for SUD remain limited and a majority of drug users relapse even after a period of treatment. The United States Food and Drug Administration (FDA) has approved several medications for opioid, nicotine, and alcohol use disorders, whereas none are approved for the treatment of cocaine or other psychostimulant use disorders. The medications approved by the FDA for the treatment of SUD can be divided into two major classes - agonist replacement therapies, such as methadone and buprenorphine for opioid use disorders (OUD), nicotine replacement therapy (NRT) and varenicline for nicotine use disorders (NUD), and antagonist therapies, such as naloxone for opioid overdose and naltrexone for promoting abstinence. In the present review, we primarily focus on the pharmacological rationale of agonist replacement strategies in treatment of opioid dependence, and the potential translation of this rationale to new therapies for cocaine use disorders. We begin by describing the neural mechanisms underlying opioid reward, followed by preclinical and clinical findings supporting the utility of agonist therapies in the treatment of OUD. We then discuss recent progress of agonist therapies for cocaine use disorders based on lessons learned from methadone and buprenorphine. We contend that future studies should identify agonist pharmacotherapies that can facilitate abstinence in patients who are motivated to quit their illicit drug use. Focusing on those that are able to achieve abstinence from cocaine will provide a platform to broaden the effectiveness of medication and psychosocial treatment strategies for this underserved population. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.
Collapse
Affiliation(s)
- Chloe J Jordan
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA.
| |
Collapse
|
20
|
Zanettini C, Scaglione A, Keighron JD, Giancola JB, Lin SC, Newman AH, Tanda G. Pharmacological classification of centrally acting drugs using EEG in freely moving rats: an old tool to identify new atypical dopamine uptake inhibitors. Neuropharmacology 2018; 161:107446. [PMID: 30481526 DOI: 10.1016/j.neuropharm.2018.11.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/05/2018] [Accepted: 11/23/2018] [Indexed: 10/27/2022]
Abstract
Atypical dopamine uptake inhibitors (DUIs) bind to the dopamine transporter and inhibit the reuptake of dopamine but have lower abuse potential than psychostimulants. Several atypical DUIs can block abuse-related effects of cocaine and methamphetamine, thus making them potential medication candidates for psychostimulant use disorders. The aim of the current study is to establish an in-vivo assay using EEG for the rapid identification of atypical DUIs with potential for medication development. The typical DUIs cocaine and methylphenidate dose-dependently decreased the power of the alpha, beta, and gamma bands. The atypical DUI modafinil and its F-analog, JBG1-049, decreased the power of beta, but in contrast to cocaine, none of the other frequency bands, while JHW007 did not significantly alter the EEG spectrum. The mu-opioid receptor agonists heroin and morphine dose-dependently decreased the power of gamma and increased power of the other bands. The effect of morphine on EEG power bands was antagonized by naltrexone. The NMDA receptor antagonist ketamine increased the power of all frequency bands. Therefore, typical and atypical DUIs and drugs of other classes differentially affected EEG spectra, showing distinctive features in the magnitude and direction of their effects on EEG. Comparative analysis of the effects of test drugs on EEG indicates a potential atypical profile of JBG1-049 with similar potency and effectiveness to its parent compound modafinil. These data suggest that EEG can be used to rapidly screen compounds for potential activity at specific pharmacological targets and provide valuable information for guiding the early stages of drug development. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
Affiliation(s)
- Claudio Zanettini
- Medication Development Program, Molecular Targets and Medications Discovery Branch, NIDA-IRP, NIH/DHHS, Baltimore, MD, USA.
| | - Alessandro Scaglione
- Neural Circuits and Cognition Unit, Laboratory of Behavioral Neuroscience, NIA-IRP, NIH/DHHS, Baltimore, MD, USA
| | - Jacqueline D Keighron
- Medication Development Program, Molecular Targets and Medications Discovery Branch, NIDA-IRP, NIH/DHHS, Baltimore, MD, USA
| | - JoLynn B Giancola
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, NIDA-IRP, NIH/DHHS, Baltimore, MD, USA
| | - Shih-Chieh Lin
- Neural Circuits and Cognition Unit, Laboratory of Behavioral Neuroscience, NIA-IRP, NIH/DHHS, Baltimore, MD, USA
| | - Amy H Newman
- Medication Development Program, Molecular Targets and Medications Discovery Branch, NIDA-IRP, NIH/DHHS, Baltimore, MD, USA; Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, NIDA-IRP, NIH/DHHS, Baltimore, MD, USA
| | - Gianluigi Tanda
- Medication Development Program, Molecular Targets and Medications Discovery Branch, NIDA-IRP, NIH/DHHS, Baltimore, MD, USA
| |
Collapse
|
21
|
Yue K, Kopajtic TA, Katz JL. Abuse liability of mitragynine assessed with a self-administration procedure in rats. Psychopharmacology (Berl) 2018; 235:2823-2829. [PMID: 30039246 DOI: 10.1007/s00213-018-4974-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/12/2018] [Indexed: 11/26/2022]
Abstract
RATIONALE Substantial use of the plant kratom for psychoactive effects has driven interest in its abuse liability. Several place conditioning studies suggest abuse liability of the active ingredient mitragynine, though studies of its self-administration have not been published. METHODS Binding of mitragynine to rat brain mu, kappa, and delta opioid receptors was compared to that for heroin and morphine. Self-administration of mitragynine, heroin, methamphetamine, or saline was assessed during single-session substitutions in rats trained to self-administer methamphetamine (0.022 mg/kg/injection, i.v.) during 1-h daily sessions. RESULTS Mitragynine had > 2- or ~ 16-fold greater affinity for the mu opioid receptor than, respectively, for kappa or delta opioid receptors. Its affinity for the mu receptor was ~ 200-fold less than that for morphine. In rats trained to self-administer methamphetamine, saline substitutions significantly decreased the number of responses, whereas different doses of methamphetamine (0.002-0.068 mg/kg/injection) or heroin (0.001-0.03 mg/kg/injection) maintained self-administration with maximal responding at 0.022 or 0.01 mg/kg/injection, respectively. In contrast, no dose of mitragynine maintained response rates greater than those obtained with saline. Presession mitragynine treatment (0.1 to 3.0 mg/kg) decreased response rates maintained by heroin but had little effect on responding maintained by methamphetamine across the same range of doses. CONCLUSIONS These results suggest a limited abuse liability of mitragynine and potential for mitragynine treatment to specifically reduce opioid abuse. With the current prevalence of opioid abuse and misuse, it appears currently that mitragynine is deserving of more extensive exploration for its development or that of an analog as a medical treatment for opioid abuse.
Collapse
Affiliation(s)
- Kai Yue
- Wuhan Institute of Biomedical Science, Jianghan University, No. 8 Sanjiaohu Street, Wuhan, China
| | - Theresa A Kopajtic
- Psychobiology Section, Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Jonathan L Katz
- Psychobiology Section, Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, 251 Bayview Blvd., Baltimore, MD, 21224, USA.
| |
Collapse
|
22
|
Hong WC, Wasko MJ, Wilkinson DS, Hiranita T, Li L, Hayashi S, Snell DB, Madura JD, Surratt CK, Katz JL. Dopamine Transporter Dynamics of N-Substituted Benztropine Analogs with Atypical Behavioral Effects. J Pharmacol Exp Ther 2018; 366:527-540. [PMID: 29945932 DOI: 10.1124/jpet.118.250498] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 06/22/2018] [Indexed: 01/07/2023] Open
Abstract
Atypical dopamine transporter (DAT) inhibitors, despite high DAT affinity, do not produce the psychomotor stimulant and abuse profile of standard DAT inhibitors such as cocaine. Proposed contributing features for those differences include off-target actions, slow onsets of action, and ligand bias regarding DAT conformation. Several 3α-(4',4''-difluoro-diphenylmethoxy)tropanes were examined, including those with the following substitutions: N-(indole-3''-ethyl)- (GA1-69), N-(R)-2''-amino-3''-methyl-n-butyl- (GA2-50), N-2''aminoethyl- (GA2-99), and N-(cyclopropylmethyl)- (JHW013). These compounds were previously reported to have rapid onset of behavioral effects and were presently evaluated pharmacologically alone or in combination with cocaine. DAT conformational mode was assessed by substituted-cysteine accessibility and molecular dynamics (MD) simulations. As determined by substituted-cysteine alkylation, all BZT analogs except GA2-99 showed bias for a cytoplasmic-facing DAT conformation, whereas cocaine stabilized the extracellular-facing conformation. MD simulations suggested that several analog-DAT complexes formed stable R85-D476 "outer gate" bonds that close the DAT to extracellular space. GA2-99 diverged from this pattern, yet had effects similar to those of other atypical DAT inhibitors. Apparent DAT association rates of the BZT analogs in vivo were slower than that for cocaine. None of the compounds was self-administered or stimulated locomotion, and each blocked those effects of cocaine. The present findings provide more detail on ligand-induced DAT conformations and indicate that aspects of DAT conformation other than "open" versus "closed" may facilitate predictions of the actions of DAT inhibitors and may promote rational design of potential treatments for psychomotor-stimulant abuse.
Collapse
Affiliation(s)
- Weimin C Hong
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, Indiana (W.C.H.); Division of Pharmaceutical Sciences (M.J.W., C.K.S.) and Department of Chemistry and Biochemistry (J.D.M.), Duquesne University, Pittsburgh; and Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland (D.S.W., T.H., L.L., S.H., D.B.S., J.L.K.)
| | - Michael J Wasko
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, Indiana (W.C.H.); Division of Pharmaceutical Sciences (M.J.W., C.K.S.) and Department of Chemistry and Biochemistry (J.D.M.), Duquesne University, Pittsburgh; and Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland (D.S.W., T.H., L.L., S.H., D.B.S., J.L.K.)
| | - Derek S Wilkinson
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, Indiana (W.C.H.); Division of Pharmaceutical Sciences (M.J.W., C.K.S.) and Department of Chemistry and Biochemistry (J.D.M.), Duquesne University, Pittsburgh; and Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland (D.S.W., T.H., L.L., S.H., D.B.S., J.L.K.)
| | - Takato Hiranita
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, Indiana (W.C.H.); Division of Pharmaceutical Sciences (M.J.W., C.K.S.) and Department of Chemistry and Biochemistry (J.D.M.), Duquesne University, Pittsburgh; and Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland (D.S.W., T.H., L.L., S.H., D.B.S., J.L.K.)
| | - Libin Li
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, Indiana (W.C.H.); Division of Pharmaceutical Sciences (M.J.W., C.K.S.) and Department of Chemistry and Biochemistry (J.D.M.), Duquesne University, Pittsburgh; and Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland (D.S.W., T.H., L.L., S.H., D.B.S., J.L.K.)
| | - Shuichiro Hayashi
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, Indiana (W.C.H.); Division of Pharmaceutical Sciences (M.J.W., C.K.S.) and Department of Chemistry and Biochemistry (J.D.M.), Duquesne University, Pittsburgh; and Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland (D.S.W., T.H., L.L., S.H., D.B.S., J.L.K.)
| | - David B Snell
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, Indiana (W.C.H.); Division of Pharmaceutical Sciences (M.J.W., C.K.S.) and Department of Chemistry and Biochemistry (J.D.M.), Duquesne University, Pittsburgh; and Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland (D.S.W., T.H., L.L., S.H., D.B.S., J.L.K.)
| | - Jeffry D Madura
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, Indiana (W.C.H.); Division of Pharmaceutical Sciences (M.J.W., C.K.S.) and Department of Chemistry and Biochemistry (J.D.M.), Duquesne University, Pittsburgh; and Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland (D.S.W., T.H., L.L., S.H., D.B.S., J.L.K.)
| | - Christopher K Surratt
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, Indiana (W.C.H.); Division of Pharmaceutical Sciences (M.J.W., C.K.S.) and Department of Chemistry and Biochemistry (J.D.M.), Duquesne University, Pittsburgh; and Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland (D.S.W., T.H., L.L., S.H., D.B.S., J.L.K.)
| | - Jonathan L Katz
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, Indiana (W.C.H.); Division of Pharmaceutical Sciences (M.J.W., C.K.S.) and Department of Chemistry and Biochemistry (J.D.M.), Duquesne University, Pittsburgh; and Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland (D.S.W., T.H., L.L., S.H., D.B.S., J.L.K.)
| |
Collapse
|
23
|
Sambo DO, Lebowitz JJ, Khoshbouei H. The sigma-1 receptor as a regulator of dopamine neurotransmission: A potential therapeutic target for methamphetamine addiction. Pharmacol Ther 2018; 186:152-167. [PMID: 29360540 PMCID: PMC5962385 DOI: 10.1016/j.pharmthera.2018.01.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Methamphetamine (METH) abuse is a major public health issue around the world, yet there are currently no effective pharmacotherapies for the treatment of METH addiction. METH is a potent psychostimulant that increases extracellular dopamine levels by targeting the dopamine transporter (DAT) and alters neuronal activity in the reward centers of the brain. One promising therapeutic target for the treatment of METH addiction is the sigma-1 receptor (σ1R). The σ1R is an endoplasmic reticulum-localized chaperone protein that is activated by cellular stress, and, unique to this chaperone, its function can also be induced or inhibited by different ligands. Upon activation of this unique "chaperone receptor", the σ1R regulates a variety of cellular functions and possesses neuroprotective activity in the brain. Interestingly, a variety of σ1R ligands modulate dopamine neurotransmission and reduce the behavioral effects of METH in animal models of addictive behavior, suggesting that the σ1R may be a viable therapeutic target for the treatment of METH addiction. In this review, we provide background on METH and the σ1R as well as a literature review regarding the role of σ1Rs in modulating both dopamine neurotransmission and the effects of METH. We aim to highlight the complexities of σ1R pharmacology and function as well as the therapeutic potential of the σ1R as a target for the treatment of METH addiction.
Collapse
Affiliation(s)
- Danielle O Sambo
- University of Florida, College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States
| | - Joseph J Lebowitz
- University of Florida, College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States
| | - Habibeh Khoshbouei
- University of Florida, College of Medicine, Department of Neuroscience, Gainesville, FL 32611, United States.
| |
Collapse
|
24
|
Dassanayake AF, Canales JJ. Replacement treatment during extinction training with the atypical dopamine uptake inhibitor, JHW-007, reduces relapse to methamphetamine seeking. Neurosci Lett 2018; 671:88-92. [PMID: 29452175 DOI: 10.1016/j.neulet.2018.02.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/26/2018] [Accepted: 02/12/2018] [Indexed: 12/29/2022]
Abstract
There are currently no approved medications to effectively counteract the effects of methamphetamine (METH), reduce its abuse and prolong abstinence from it. Data accumulated in recent years have shown that a range of N-substituted benztropine (BZT) analogues possesses psychopharmacological features consistent with those of a potential replacement or "substitute" treatment for stimulant addiction. On the other hand, the evidence that antidepressant therapy may effectively prevent relapse to stimulant seeking is controversial. Here, we compared in rats the ability of the BZT analogue and high affinity dopamine (DA) reuptake inhibitor, JHW-007, and the antidepressant, trazodone, administered during extinction sessions after chronic METH self-administration, to alter METH-primed reinstatement of drug seeking. The data showed that trazodone produced paradoxical effects on lever pressing during extinction of METH self-administration, decreasing active, but increasing inactive, lever pressing. JHW-007 did not have any observable effects on extinction training. Importantly, JHW-007 significantly attenuated METH-primed reinstatement, whereas trazodone enhanced it. These findings lend support to the candidacy of selective DA uptake blockers, such as JHW-007, as potential treatments for METH addiction, but not to the use of antidepressant medication as a single therapeutic approach for relapse prevention.
Collapse
Affiliation(s)
- Ashlea F Dassanayake
- Division of Psychology, School of Medicine, University of Tasmania, Private Bag 30, Hobart, TAS, 7001, Australia
| | - Juan J Canales
- Division of Psychology, School of Medicine, University of Tasmania, Private Bag 30, Hobart, TAS, 7001, Australia.
| |
Collapse
|
25
|
Behavioral economic analysis of the effects of N-substituted benztropine analogs on cocaine self-administration in rats. Psychopharmacology (Berl) 2018; 235:47-58. [PMID: 28932889 DOI: 10.1007/s00213-017-4739-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 09/12/2017] [Indexed: 10/18/2022]
Abstract
RATIONALE AND OBJECTIVES Benztropine (BZT) analogs and other atypical dopamine uptake inhibitors selectively decrease cocaine self-administration at doses that do not affect responding maintained by other reinforcers. Those effects were further characterized in the current study using a behavioral economic assessment of how response requirement (price) affects reinforcers obtained (consumption) in rats. METHODS Two groups of rats were trained to press levers with food (45-mg pellet) or cocaine (0.32 mg/kg/injection) reinforcement under fixed-ratio (FR) 5-response schedules. In selected sessions, the FR requirement was increased (5-80) during successive 20-min components to determine demand curves, which plot consumption against price. An exponential function was fitted to the data to derive the consumption at zero price (Q 0) and the rate of decrease in consumption (essential value, EV) with increased price. The BZT analogs, AHN1-055, AHN2-005, JHW007 (3.2-10 or 17.8 mg/kg, each), vehicle, or comparison drugs (methylphenidate, ketamine), were administered i.p. before selected demand-curve determinations. RESULTS Consumption of cocaine or food decreased with increased FR requirement. Each drug shifted the demand curve rightward at the lowest doses and leftward/downward at higher doses. The effects on EV and Q 0 were greater for cocaine than for food-reinforced responding. Additionally, the effects of the BZT analogs on EV and Q 0 were greater than those obtained with a standard dopamine transport inhibitor, methylphenidate, and the NMDA antagonist, ketamine (1.0-10.0 mg/kg, each). With these latter drugs, the demand-curve parameters were affected similarly with cocaine and food-maintained responding. CONCLUSIONS The current findings, obtained using a behavioral economic assessment, suggest that BZT analogs selectively decrease the reinforcing effectiveness of cocaine.
Collapse
|
26
|
Sambo DO, Lin M, Owens A, Lebowitz JJ, Richardson B, Jagnarine DA, Shetty M, Rodriquez M, Alonge T, Ali M, Katz J, Yan L, Febo M, Henry LK, Bruijnzeel AW, Daws L, Khoshbouei H. The sigma-1 receptor modulates methamphetamine dysregulation of dopamine neurotransmission. Nat Commun 2017; 8:2228. [PMID: 29263318 PMCID: PMC5738444 DOI: 10.1038/s41467-017-02087-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 11/05/2017] [Indexed: 12/16/2022] Open
Abstract
Dopamine neurotransmission is highly dysregulated by the psychostimulant methamphetamine, a substrate for the dopamine transporter (DAT). Through interactions with DAT, methamphetamine increases extracellular dopamine levels in the brain, leading to its rewarding and addictive properties. Methamphetamine also interacts with the sigma-1 receptor (σ1R), an inter-organelle signaling modulator. Using complementary strategies, we identified a novel mechanism for σ1R regulation of dopamine neurotransmission in response to methamphetamine. We found that σ1R activation prevents methamphetamine-induced, DAT-mediated increases in firing activity of dopamine neurons. In vitro and in vivo amperometric measurements revealed that σ1R activation decreases methamphetamine-stimulated dopamine efflux without affecting basal dopamine neurotransmission. Consistent with these findings, σ1R activation decreases methamphetamine-induced locomotion, motivated behavior, and enhancement of brain reward function. Notably, we revealed that the σ1R interacts with DAT at or near the plasma membrane and decreases methamphetamine-induced Ca2+ signaling, providing potential mechanisms. Broadly, these data provide evidence for σ1R regulation of dopamine neurotransmission and support the σ1R as a putative target for the treatment of methamphetamine addiction.
Collapse
Affiliation(s)
- Danielle O Sambo
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Min Lin
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Anthony Owens
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Joseph J Lebowitz
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Ben Richardson
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Darin A Jagnarine
- Department of Psychiatry, University of Florida, Gainesville, FL, 32611, USA
| | - Madhur Shetty
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Meghan Rodriquez
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Taiwo Alonge
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Mishaal Ali
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA
| | - Jonathan Katz
- Psychobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Long Yan
- Max Plank Institute for Neuroscience Jupiter, Jupiter, FL, 33458, USA
| | - Marcelo Febo
- Department of Psychiatry, University of Florida, Gainesville, FL, 32611, USA
| | - L Keith Henry
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | | | - Lynette Daws
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
27
|
Tunstall BJ, Ho CP, Cao J, Vendruscolo JCM, Schmeichel BE, Slack RD, Tanda G, Gadiano AJ, Rais R, Slusher BS, Koob GF, Newman AH, Vendruscolo LF. Atypical dopamine transporter inhibitors attenuate compulsive-like methamphetamine self-administration in rats. Neuropharmacology 2017; 131:96-103. [PMID: 29217282 DOI: 10.1016/j.neuropharm.2017.12.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 11/30/2022]
Abstract
Methamphetamine (METH) is a highly addictive drug, but no pharmacological treatment is yet available for METH use disorders. Similar to METH, the wake-promoting drug (R)-modafinil (R-MOD) binds to the dopamine transporter (DAT). Unlike METH, R-MOD is not a substrate for transport by DAT and has low abuse potential. We tested the hypothesis that the atypical DAT inhibitor R-MOD and compounds that are derived from modafinil would decrease METH intake by reducing the actions of METH at the DAT. We tested the effects of systemic injections of R-MOD and four novel modafinil-derived ligands with increased DAT affinity (JJC8-016, JJC8-088, JJC8-089, and JJC8-091) on intravenous (i.v.) METH self-administration in rats that were allowed short access (ShA; 1 h) or long access (LgA; 6 h) to the drug. ShA rats exhibited stable METH intake over sessions, whereas LgA rats exhibited an escalation of drug intake. R-MOD decreased METH self-administration in ShA and LgA rats (in the 1st hour only). JJC8-091 and JJC8-016 decreased METH self-administration in both ShA and LgA rats. JJC8-089 decreased METH self-administration in LgA rats only, whereas JJC8-088 had no effect on METH self-administration in either ShA or LgA rats. These findings support the potential of atypical DAT inhibitors for the treatment of METH use disorders and suggest several novel compounds as candidate drugs.
Collapse
Affiliation(s)
- Brendan J Tunstall
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Chelsea P Ho
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Janaína C M Vendruscolo
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Brooke E Schmeichel
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Rachel D Slack
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Gianluigi Tanda
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Alexandra J Gadiano
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA; Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rana Rais
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barbara S Slusher
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - George F Koob
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Amy H Newman
- Molecular Targets and Medications Discovery Program, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA.
| | - Leandro F Vendruscolo
- Neurobiology of Addiction Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
28
|
Hofford RS, Chow JJ, Beckmann JS, Bardo MT. Effects of environmental enrichment on self-administration of the short-acting opioid remifentanil in male rats. Psychopharmacology (Berl) 2017; 234:3499-3506. [PMID: 28916995 PMCID: PMC6541008 DOI: 10.1007/s00213-017-4734-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 09/05/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND Opioid abuse is a major problem around the world. Identifying environmental factors that contribute to opioid abuse and addiction is necessary for decreasing this epidemic. In rodents, environmental enrichment protects against the development of low dose stimulant self-administration, but studies examining the effect of enrichment and isolation (compared to standard housing) on the development of intravenous opioid self-administration have not been conducted. The present study investigated the role of environmental enrichment on self-administration of the short-acting μ-opioid remifentanil. METHODS Rats were raised in an enriched condition (Enr), standard condition (Std), or isolated condition (Iso) beginning at 21 days of age and were trained to lever press for 1 or 3 μg/kg/infusion remifentanil in young adulthood. Acquisition of self-administration and responding during increasing fixed ratio requirements were assessed, and a dose-response curve was generated. RESULTS In all phases, Enr rats lever pressed significantly less than Std and Iso rats, with Enr rats pressing between 9 and 40% the amount of Iso rats. Enr rats did not acquire remifentanil self-administration when trained with 1 μg/kg/infusion, did not increase responding over increasing FR when trained at either dose, and their dose-response curves were flattened compared to Std and Iso rats. When expressed as economic demand curves, Enr rats displayed a decrease in both essential value (higher α) and reinforcer intensity (Q 0) compared to Std and Iso rats at the 1 μg/kg/infusion training dose. CONCLUSION Environmental enrichment reduced remifentanil intake, suggesting that social and environmental novelty may protect against opioid abuse.
Collapse
Affiliation(s)
- Rebecca S Hofford
- Department of Psychology, BBSRB, University of Kentucky, Room 448C, Lexington, KY, 40536-0509, USA.
| | - Jonathan J Chow
- Department of Psychology, BBSRB, University of Kentucky, Room 448C, Lexington, KY, 40536-0509, USA
| | - Joshua S Beckmann
- Department of Psychology, BBSRB, University of Kentucky, Room 448C, Lexington, KY, 40536-0509, USA
| | - Michael T Bardo
- Department of Psychology, BBSRB, University of Kentucky, Room 448C, Lexington, KY, 40536-0509, USA
| |
Collapse
|
29
|
Avelar AJ, Cao J, Newman AH, Beckstead MJ. Atypical dopamine transporter inhibitors R-modafinil and JHW 007 differentially affect D2 autoreceptor neurotransmission and the firing rate of midbrain dopamine neurons. Neuropharmacology 2017; 123:410-419. [PMID: 28625719 PMCID: PMC5546153 DOI: 10.1016/j.neuropharm.2017.06.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/17/2017] [Accepted: 06/14/2017] [Indexed: 12/12/2022]
Abstract
Abuse of psychostimulants like cocaine that inhibit dopamine (DA) reuptake through the dopamine transporter (DAT) represents a major public health issue, however FDA-approved pharmacotherapies have yet to be developed. Recently a class of ligands termed "atypical DAT inhibitors" has gained attention due to their range of effectiveness in increasing extracellular DA levels without demonstrating significant abuse liability. These compounds not only hold promise as therapeutic agents to treat stimulant use disorders but also as experimental tools to improve our understanding of DAT function. Here we used patch clamp electrophysiology in mouse brain slices to explore the effects of two atypical DAT inhibitors (R-modafinil and JHW 007) on the physiology of single DA neurons in the substantia nigra and ventral tegmental area. Despite their commonalities of being DAT inhibitors that lack cocaine-like behavioral profiles, these compounds exhibited surprisingly divergent cellular effects. Similar to cocaine, R-modafinil slowed DA neuron firing in a D2 receptor-dependent manner and rapidly enhanced the amplitude and duration of D2 receptor-mediated currents in the midbrain. In contrast, JHW 007 exhibited little effect on firing, slow DAT blockade, and an unexpected inhibition of D2 receptor-mediated currents that may be due to direct D2 receptor antagonism. Furthermore, pretreatment with JHW 007 blunted the cellular effects of cocaine, suggesting that it may be valuable to investigate similar DAT inhibitors as potential therapeutic agents. Further exploration of these and other atypical DAT inhibitors may reveal important cellular effects of compounds that will have potential as pharmacotherapies for treating cocaine use disorders.
Collapse
Affiliation(s)
- Alicia J Avelar
- Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX, 78229, USA.
| | - Jianjing Cao
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| | - Michael J Beckstead
- Department of Cellular and Integrative Physiology, UT Health Science Center, San Antonio, TX, 78229, USA.
| |
Collapse
|
30
|
Hiranita T, Hong WC, Kopajtic T, Katz JL. σ Receptor Effects of N-Substituted Benztropine Analogs: Implications for Antagonism of Cocaine Self-Administration. J Pharmacol Exp Ther 2017; 362:2-13. [PMID: 28442581 PMCID: PMC5454590 DOI: 10.1124/jpet.117.241109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/20/2017] [Indexed: 11/22/2022] Open
Abstract
Several N-substituted benztropine (BZT) analogs are atypical dopamine transport inhibitors as they have affinity for the dopamine transporter (DAT) but have minimal cocaine-like pharmacologic effects and can block numerous effects of cocaine, including its self-administration. Among these compounds, N-methyl (AHN1-055), N-allyl (AHN2-005), and N-butyl (JHW007) analogs of 3α-[bis(4'-fluorophenyl)methoxy]-tropane were more potent in antagonizing self-administration of cocaine and d-methamphetamine than in decreasing food-maintained responding. The antagonism of cocaine self-administration (0.03-1.0 mg/kg per injection) with the above BZT analogs was reproduced in the present study. Further, the stimulant-antagonist effects resembled previously reported effects of pretreatments with combinations of standard DAT inhibitors and σ1-receptor (σ1R) antagonists. Therefore, the present study examined binding of the BZT analogs to σRs, as well as their in vivo σR antagonist effects. Each of the BZT analogs displaced radiolabeled σR ligands with nanomolar affinity. Further, self-administration of the σR agonist DTG (0.1-3.2 mg/kg/injection) was dose dependently blocked by AHN2-005 and JHW007 but potentiated by AHN1-055. In contrast, none of the BZT analogs that were active against DTG self-administration was active against the self-administration of agonists at dopamine D1-like [R(+)-SKF 81297, (±)-SKF 82958 (0.00032-0.01 mg/kg per injection each)], D2-like [R(-)-NPA (0.0001-0.0032 mg/kg per injection), (-)-quinpirole (0.0032-0.1 mg/kg per injection)], or μ-opioid (remifentanil, 0.0001-0.0032 mg/kg per injection) receptors. The present results indicate that behavioral antagonist effects of the N-substituted BZT analogs are specific for abused drugs acting at the DAT and further suggest that σR antagonism contributes to those actions.
Collapse
Affiliation(s)
- Takato Hiranita
- Psychobiology Section, Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health (T.H., T.K., J.L.K.), and Department of Pharmaceutical Sciences, Butler University (W.C.H.), Indianapolis, Indiana
| | - Weimin C Hong
- Psychobiology Section, Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health (T.H., T.K., J.L.K.), and Department of Pharmaceutical Sciences, Butler University (W.C.H.), Indianapolis, Indiana
| | - Theresa Kopajtic
- Psychobiology Section, Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health (T.H., T.K., J.L.K.), and Department of Pharmaceutical Sciences, Butler University (W.C.H.), Indianapolis, Indiana
| | - Jonathan L Katz
- Psychobiology Section, Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health (T.H., T.K., J.L.K.), and Department of Pharmaceutical Sciences, Butler University (W.C.H.), Indianapolis, Indiana
| |
Collapse
|
31
|
Relapse to cocaine seeking in an invertebrate. Pharmacol Biochem Behav 2017; 157:41-46. [PMID: 28455125 DOI: 10.1016/j.pbb.2017.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/07/2017] [Accepted: 04/21/2017] [Indexed: 12/21/2022]
Abstract
Addiction is characterised by cycles of compulsive drug taking, periods of abstinence and episodes of relapse. The extinction/reinstatement paradigm has been extensively used in rodents to model human relapse and explore underlying mechanisms and therapeutics. However, relapse to drug seeking behaviour has not been previously demonstrated in invertebrates. Here, we used a cocaine conditioned place preference (CPP) paradigm in the flatworm, planarian, followed by extinction and reinstatement of drug seeking. Once baseline preference was established for one of two distinctly textured environments (i.e. compartments with a coarse or smooth surface), planarian received pairings of cocaine (5μM) in the non-preferred, and vehicle in the most preferred, environment, and were tested for conditioning thereafter. Cocaine produced robust CPP, measured as a significant increase in the time spent in the cocaine-paired compartment. Subsequently, planarian underwent extinction training, reverting back to their original preference within three sessions. Brief exposure to cocaine (5μM) or methamphetamine (5μM) reinstated cocaine-seeking behaviour. By contrast, the high affinity dopamine transporter inhibitor, (N-(n-butyl)-3α-[bis (4-fluorophenyl) methoxy]-tropane) (JHW007), which in rodents exhibits a neurochemical and behavioural profile distinct from cocaine, was ineffective. The present findings demonstrate for the first time reinstatement of extinguished cocaine seeking in an invertebrate model and suggest that the long-term adaptations underlying drug conditioning and relapse are highly conserved through evolution.
Collapse
|
32
|
Behavioral evidence for the abuse potential of the novel synthetic cathinone alpha-pyrrolidinopentiothiophenone (PVT) in rodents. Psychopharmacology (Berl) 2017; 234:857-867. [PMID: 28070621 DOI: 10.1007/s00213-017-4526-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/29/2016] [Indexed: 12/31/2022]
Abstract
RATIONALE Synthetic cathinones are chemical derivatives of cathinone that are pharmacologically similar to cocaine and methamphetamine. Recently, abuse of synthetic cathinones among young people has increased. OBJECTIVES The present study aimed to characterize the behavioral effects of alpha-pyrrolidinopentiothiophenone (PVT), an analog of alpha-pyrrolidinovalerophenone and second-generation synthetic cathinone, as well as to evaluate its abuse potential, using conditioned place preference, intravenous self-administration (SA), and drug discrimination paradigms in rodent models. RESULTS Alpha-PVT produced a significant place preference in mice at doses of 10, 30, and 50 mg/kg. In the SA experiment, alpha-PVT (0.1, 0.3, and 1.0 mg/kg/infusion) produced an inverted U-shaped dose-effect curve in rats. Under a progressive ratio schedule of reinforcement, there appeared to be a positive relationship between alpha-PVT dose and the breakpoints for alpha-PVT reinforcement. Additionally, alpha-PVT fully substituted for the discriminative stimulus effects of both cocaine and methamphetamine in rats. CONCLUSIONS Our results indicate that alpha-PVT has rewarding and reinforcing effects and shares the interoceptive effects of cocaine and methamphetamine. To the best of our knowledge, the present study is the first to show that alpha-PVT has reinforcing properties when delivered on its own, which suggests possible abuse liability in humans.
Collapse
|
33
|
Abstract
This paper is the thirty-eighth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2015 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia, stress and social status, tolerance and dependence, learning and memory, eating and drinking, drug abuse and alcohol, sexual activity and hormones, pregnancy, development and endocrinology, mental illness and mood, seizures and neurologic disorders, electrical-related activity and neurophysiology, general activity and locomotion, gastrointestinal, renal and hepatic functions, cardiovascular responses, respiration and thermoregulation, and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
34
|
Katz JL, Hiranita T, Hong WC, Job MO, McCurdy CR. A Role for Sigma Receptors in Stimulant Self-Administration and Addiction. Handb Exp Pharmacol 2017; 244:177-218. [PMID: 28110353 DOI: 10.1007/164_2016_94] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Sigma receptors (σRs) are structurally unique proteins that function intracellularly as chaperones. Historically, σRs have been implicated as modulators of psychomotor stimulant effects and have at times been proposed as potential avenues for modifying stimulant abuse. However, the influence of ligands for σRs on the effects of stimulants, such as cocaine or methamphetamine, in various preclinical procedures related to drug abuse has been varied. The present paper reviews the effects of σR agonists and antagonists in three particularly relevant procedures: stimulant discrimination, place conditioning, and self-administration. The literature to date suggests limited σR involvement in the discriminative-stimulus effects of psychomotor stimulants, either with σR agonists substituting for the stimulant or with σR antagonists blocking stimulant effects. In contrast, studies of place conditioning suggest that administration of σR antagonists or down-regulation of σR protein can block the place conditioning induced by stimulants. Despite place conditioning results, selective σR antagonists are inactive in blocking the self-administration of stimulants. However, compounds binding to the dopamine transporter and blocking σRs can selectively decrease stimulant self-administration. Further, after self-administration of stimulants, σR agonists are self-administered, an effect not seen in subjects without that specific history. These findings suggest that stimulants induce unique changes in σR activity, and once established, the changes induced create redundant, and dopamine independent reinforcement pathways. Concomitant targeting of both dopaminergic pathways and σR proteins produces a selective antagonism of those pathways, suggesting new avenues for combination chemotherapies to specifically combat stimulant abuse.
Collapse
Affiliation(s)
- Jonathan L Katz
- Psychobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD, 21224, USA.
| | - Takato Hiranita
- Psychobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD, 21224, USA
| | - Weimin C Hong
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, IN, 46208, USA
| | - Martin O Job
- Psychobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD, 21224, USA
| | - Christopher R McCurdy
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, 38677, USA
| |
Collapse
|
35
|
Hiranita T, Freyberg Z. Importance of Substrate-Coupled Proton Antiport by the Vesicular Monoamine Transporter in the Actions of Amphetamines in Drosophila Brain. ACTA ACUST UNITED AC 2016; 4. [PMID: 28164142 DOI: 10.4172/2329-6488.1000e136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Takato Hiranita
- Division of Neurotoxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), AR 72079-9502, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
36
|
Abstract
Sigma-1 receptors (σ1Rs) are structurally unique intracellular proteins that function as chaperones. σ1Rs translocate from the mitochondria-associated membrane to other subcellular compartments, and can influence a host of targets, including ion channels, G-protein-coupled receptors, lipids, and other signaling proteins. Drugs binding to σRs can induce or block the actions of σRs. Studies indicate that stimulant self-administration induces the reinforcing effects of σR agonists, because of dopamine transporter actions. Once established, the reinforcing effects of σR agonists are independent of dopaminergic mechanisms traditionally thought to be critical to the reinforcing effects of stimulants. Self-administered doses of σR agonists do not increase dopamine concentrations in the nucleus accumbens shell, a transmitter and brain region considered important for the reinforcing effects of abused drugs. However, self-administration of σR agonists is blocked by σR antagonists. Several effects of stimulants have been blocked by σR antagonists, including the reinforcing effects, assessed by a place-conditioning procedure. However, the self-administration of stimulants is largely unaffected by σR antagonists, indicating fundamental differences in the mechanisms underlying these two procedures used to assess the reinforcing effects. When σR antagonists are administered in combination with dopamine uptake inhibitors, an effective and specific blockade of stimulant self-administration is obtained. Actions of stimulant drugs related to their abuse induce unique changes in σR activity and the changes induced potentially create redundant and, once established, independent reinforcement pathways. Concomitant targeting of both dopaminergic pathways and σR proteins produces a selective antagonism of stimulant self-administration, suggesting new avenues for combination chemotherapies to specifically combat stimulant abuse.
Collapse
|
37
|
Banks ML. Utility of preclinical drug versus food choice procedures to evaluate candidate medications for methamphetamine use disorder. Ann N Y Acad Sci 2016; 1394:92-105. [PMID: 27936284 DOI: 10.1111/nyas.13276] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/01/2016] [Accepted: 09/16/2016] [Indexed: 01/21/2023]
Abstract
Substance use disorders are diagnosed as a manifestation of inappropriate behavioral allocation toward abused drugs and away from other behaviors maintained by more adaptive nondrug reinforcers (e.g., money and social relationships). Substance use disorder treatment goals include not only decreasing drug-maintained behavior but also promoting behavioral reallocation toward these socially adaptive alternative reinforcers. Preclinical drug self-administration procedures that offer concurrent access to both drug and nondrug reinforcers provide a translationally relevant dependent measure of behavioral allocation that may be useful for candidate medication evaluation. In contrast to other abused drugs, such as heroin or cocaine, preclinical methamphetamine versus food choice procedures have been a more recent development. We hypothesize that preclinical to clinical translatability would be improved by the evaluation of repeated pharmacological treatment effects on methamphetamine self-administration under a methamphetamine versus food choice procedure. In support of this hypothesis, a literature review suggests strong concordance between preclinical pharmacological treatment effects on methamphetamine versus food choice in nonhuman primates and clinical medication treatment effects on methamphetamine self-administration in human laboratory studies or methamphetamine abuse metrics in clinical trials. In conclusion, this literature suggests preclinical methamphetamine versus food choice procedures may be useful in developing innovative pharmacotherapies for methamphetamine use disorder.
Collapse
Affiliation(s)
- Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
38
|
Di Giovanni G, Svob Strac D, Sole M, Unzeta M, Tipton KF, Mück-Šeler D, Bolea I, Della Corte L, Nikolac Perkovic M, Pivac N, Smolders IJ, Stasiak A, Fogel WA, De Deurwaerdère P. Monoaminergic and Histaminergic Strategies and Treatments in Brain Diseases. Front Neurosci 2016; 10:541. [PMID: 27932945 PMCID: PMC5121249 DOI: 10.3389/fnins.2016.00541] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 11/07/2016] [Indexed: 12/18/2022] Open
Abstract
The monoaminergic systems are the target of several drugs for the treatment of mood, motor and cognitive disorders as well as neurological conditions. In most cases, advances have occurred through serendipity, except for Parkinson's disease where the pathophysiology led almost immediately to the introduction of dopamine restoring agents. Extensive neuropharmacological studies first showed that the primary target of antipsychotics, antidepressants, and anxiolytic drugs were specific components of the monoaminergic systems. Later, some dramatic side effects associated with older medicines were shown to disappear with new chemical compounds targeting the origin of the therapeutic benefit more specifically. The increased knowledge regarding the function and interaction of the monoaminergic systems in the brain resulting from in vivo neurochemical and neurophysiological studies indicated new monoaminergic targets that could achieve the efficacy of the older medicines with fewer side-effects. Yet, this accumulated knowledge regarding monoamines did not produce valuable strategies for diseases where no monoaminergic drug has been shown to be effective. Here, we emphasize the new therapeutic and monoaminergic-based strategies for the treatment of psychiatric diseases. We will consider three main groups of diseases, based on the evidence of monoamines involvement (schizophrenia, depression, obesity), the identification of monoamines in the diseases processes (Parkinson's disease, addiction) and the prospect of the involvement of monoaminergic mechanisms (epilepsy, Alzheimer's disease, stroke). In most cases, the clinically available monoaminergic drugs induce widespread modifications of amine tone or excitability through neurobiological networks and exemplify the overlap between therapeutic approaches to psychiatric and neurological conditions. More recent developments that have resulted in improved drug specificity and responses will be discussed in this review.
Collapse
Affiliation(s)
| | | | - Montse Sole
- Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Institut de Neurociències, Universitat Autònoma de BarcelonaBarcelona, Spain
| | - Mercedes Unzeta
- Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Institut de Neurociències, Universitat Autònoma de BarcelonaBarcelona, Spain
| | - Keith F. Tipton
- School of Biochemistry and Immunology, Trinity College DublinDublin, Ireland
| | - Dorotea Mück-Šeler
- Division of Molecular Medicine, Rudjer Boskovic InstituteZagreb, Croatia
| | - Irene Bolea
- Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Institut de Neurociències, Universitat Autònoma de BarcelonaBarcelona, Spain
| | | | | | - Nela Pivac
- Division of Molecular Medicine, Rudjer Boskovic InstituteZagreb, Croatia
| | - Ilse J. Smolders
- Department of Pharmaceutical Chemistry and Drug Analysis, Vrije Universiteit BrusselBrussels, Belgium
| | - Anna Stasiak
- Department of Hormone Biochemistry, Medical University of LodzLodz, Poland
| | - Wieslawa A. Fogel
- Department of Hormone Biochemistry, Medical University of LodzLodz, Poland
| | - Philippe De Deurwaerdère
- Centre National de la Recherche Scientifique (Unité Mixte de Recherche 5293), Institut of Neurodegenerative DiseasesBordeaux Cedex, France
| |
Collapse
|
39
|
Golovko AI, Bonitenko EY, Ivanov MB, Barinov VA, Zatsepin EP. The neurochemical bases of the pharmacological activity of ligands of monoamine-transport systems. NEUROCHEM J+ 2016. [DOI: 10.1134/s1819712416030065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
40
|
German CL, Baladi MG, McFadden LM, Hanson GR, Fleckenstein AE. Regulation of the Dopamine and Vesicular Monoamine Transporters: Pharmacological Targets and Implications for Disease. Pharmacol Rev 2016; 67:1005-24. [PMID: 26408528 DOI: 10.1124/pr.114.010397] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dopamine (DA) plays a well recognized role in a variety of physiologic functions such as movement, cognition, mood, and reward. Consequently, many human disorders are due, in part, to dysfunctional dopaminergic systems, including Parkinson's disease, attention deficit hyperactivity disorder, and substance abuse. Drugs that modify the DA system are clinically effective in treating symptoms of these diseases or are involved in their manifestation, implicating DA in their etiology. DA signaling and distribution are primarily modulated by the DA transporter (DAT) and by vesicular monoamine transporter (VMAT)-2, which transport DA into presynaptic terminals and synaptic vesicles, respectively. These transporters are regulated by complex processes such as phosphorylation, protein-protein interactions, and changes in intracellular localization. This review provides an overview of 1) the current understanding of DAT and VMAT2 neurobiology, including discussion of studies ranging from those conducted in vitro to those involving human subjects; 2) the role of these transporters in disease and how these transporters are affected by disease; and 3) and how selected drugs alter the function and expression of these transporters. Understanding the regulatory processes and the pathologic consequences of DAT and VMAT2 dysfunction underlies the evolution of therapeutic development for the treatment of DA-related disorders.
Collapse
Affiliation(s)
- Christopher L German
- School of Dentistry (C.L.G., M.G.B., G.R.H., A.E.F.) and Department of Pharmacology and Toxicology (L.M.M., G.R.H.), University of Utah, Salt Lake City, Utah
| | - Michelle G Baladi
- School of Dentistry (C.L.G., M.G.B., G.R.H., A.E.F.) and Department of Pharmacology and Toxicology (L.M.M., G.R.H.), University of Utah, Salt Lake City, Utah
| | - Lisa M McFadden
- School of Dentistry (C.L.G., M.G.B., G.R.H., A.E.F.) and Department of Pharmacology and Toxicology (L.M.M., G.R.H.), University of Utah, Salt Lake City, Utah
| | - Glen R Hanson
- School of Dentistry (C.L.G., M.G.B., G.R.H., A.E.F.) and Department of Pharmacology and Toxicology (L.M.M., G.R.H.), University of Utah, Salt Lake City, Utah
| | - Annette E Fleckenstein
- School of Dentistry (C.L.G., M.G.B., G.R.H., A.E.F.) and Department of Pharmacology and Toxicology (L.M.M., G.R.H.), University of Utah, Salt Lake City, Utah
| |
Collapse
|
41
|
Katz JL, Hiranita T, Kopajtic TA, Rice KC, Mesangeau C, Narayanan S, Abdelazeem AH, McCurdy CR. Blockade of Cocaine or σ Receptor Agonist Self Administration by Subtype-Selective σ Receptor Antagonists. J Pharmacol Exp Ther 2016; 358:109-24. [PMID: 27189970 DOI: 10.1124/jpet.116.232728] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/14/2016] [Indexed: 11/22/2022] Open
Abstract
The identification of sigma receptor (σR) subtypes has been based on radioligand binding and, despite progress with σ1R cellular function, less is known about σR subtype functions in vivo. Recent findings that cocaine self administration experience will trigger σR agonist self administration was used in this study to assess the in vivo receptor subtype specificity of the agonists (+)-pentazocine, PRE-084 [2-(4-morpholinethyl) 1-phenylcyclohexanecarboxylate hydrochloride], and 1,3-di-o-tolylguanidine (DTG) and several novel putative σR antagonists. Radioligand binding studies determined in vitro σR selectivity of the novel compounds, which were subsequently studied for self administration and antagonism of cocaine, (+)-pentazocine, PRE-084, or DTG self administration. Across the dose ranges studied, none of the novel compounds were self administered, nor did they alter cocaine self administration. All compounds blocked DTG self administration, with a subset also blocking (+)-pentazocine and PRE-084 self administration. The most selective of the compounds in binding σ1Rs blocked cocaine self administration when combined with a dopamine transport inhibitor, either methylphenidate or nomifensine. These drug combinations did not decrease rates of responding maintained by food reinforcement. In contrast, the most selective of the compounds in binding σ2Rs had no effect on cocaine self administration in combination with either dopamine transport inhibitor. Thus, these results identify subtype-specific in vivo antagonists, and the utility of σR agonist substitution for cocaine self administration as an assay capable of distinguishing σR subtype selectivity in vivo. These results further suggest that effectiveness of dual σR antagonism and dopamine transport inhibition in blocking cocaine self administration is specific for σ1Rs and further support this dual targeting approach to development of cocaine antagonists.
Collapse
Affiliation(s)
- Jonathan L Katz
- Psychobiology Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse, Baltimore, Maryland (J.L.K., T.H., T.A.K.); Drug Design and Synthesis Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Baltimore, Maryland (K.C.R.); and Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi (C.M., S.N., A.H.A., C.R.M.)
| | - Takato Hiranita
- Psychobiology Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse, Baltimore, Maryland (J.L.K., T.H., T.A.K.); Drug Design and Synthesis Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Baltimore, Maryland (K.C.R.); and Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi (C.M., S.N., A.H.A., C.R.M.)
| | - Theresa A Kopajtic
- Psychobiology Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse, Baltimore, Maryland (J.L.K., T.H., T.A.K.); Drug Design and Synthesis Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Baltimore, Maryland (K.C.R.); and Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi (C.M., S.N., A.H.A., C.R.M.)
| | - Kenner C Rice
- Psychobiology Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse, Baltimore, Maryland (J.L.K., T.H., T.A.K.); Drug Design and Synthesis Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Baltimore, Maryland (K.C.R.); and Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi (C.M., S.N., A.H.A., C.R.M.)
| | - Christophe Mesangeau
- Psychobiology Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse, Baltimore, Maryland (J.L.K., T.H., T.A.K.); Drug Design and Synthesis Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Baltimore, Maryland (K.C.R.); and Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi (C.M., S.N., A.H.A., C.R.M.)
| | - Sanju Narayanan
- Psychobiology Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse, Baltimore, Maryland (J.L.K., T.H., T.A.K.); Drug Design and Synthesis Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Baltimore, Maryland (K.C.R.); and Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi (C.M., S.N., A.H.A., C.R.M.)
| | - Ahmed H Abdelazeem
- Psychobiology Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse, Baltimore, Maryland (J.L.K., T.H., T.A.K.); Drug Design and Synthesis Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Baltimore, Maryland (K.C.R.); and Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi (C.M., S.N., A.H.A., C.R.M.)
| | - Christopher R McCurdy
- Psychobiology Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse, Baltimore, Maryland (J.L.K., T.H., T.A.K.); Drug Design and Synthesis Section, Intramural Research Program, National Institutes of Health National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, Baltimore, Maryland (K.C.R.); and Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, University, Mississippi (C.M., S.N., A.H.A., C.R.M.)
| |
Collapse
|
42
|
Yasui Y, Su TP. Potential Molecular Mechanisms on the Role of the Sigma-1 Receptor in the Action of Cocaine and Methamphetamine. ACTA ACUST UNITED AC 2016; 5. [PMID: 27088037 DOI: 10.4303/jdar/235970] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The sigma-1 receptor (Sig-1R) is an endoplasmic reticulum membrane protein that involves a wide range of physiological functions. The Sig-1R has been shown to bind psychostimulants including cocaine and methamphetamine (METH) and thus has been implicated in the actions of those psychostimulants. For example, it has been demonstrated that the Sig-1R antagonists mitigate certain behavioral and cellular effects of psychostimulants including hyperactivity and neurotoxicity. Thus, the Sig-1R has become a potential therapeutic target of medication development against drug abuse that differs from traditional monoamine-related strategies. In this review, we will focus on the molecular mechanisms of the Sig-1R and discuss in such a manner with a hope to further understand or unveil unexplored relations between the Sig-1R and the actions of cocaine and METH, particularly in the context of cellular biological relevance.
Collapse
Affiliation(s)
- Yuko Yasui
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, Baltimore, Maryland 21224
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, Baltimore, Maryland 21224
| |
Collapse
|
43
|
Hiranita T, Collins GT. Differential Roles for Dopamine D1-Like and D2-Like Receptors in Mediating the Reinforcing Effects of Cocaine: Convergent Evidence from Pharmacological and Genetic Studies. ACTA ACUST UNITED AC 2015; 3. [PMID: 27390753 PMCID: PMC4932862 DOI: 10.4172/2329-6488.1000e124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A series of studies by Drs. Barak Caine, James Woods, Gregory Collins, Jonathan Katz and Takato Hiranita demonstrated a novel and unique reinforcing effect using dopamine (DA) D2-like receptor [D2-like R: D2, D3, and D4 receptor subtypes (respectively, D2R, D3R, and D4R)] agonists in rats and genetically modified mice. In order to understand how important their findings are, a comparison was made regarding the reinforcing effects of DA D2-like R full agonists with those of DA uptake inhibitors and of a DA D1-like receptor [D1-like R, D1 and D5 receptor subtypes (D1R and D5R)] full agonist (±)-SKF 82958.
Collapse
Affiliation(s)
- Takato Hiranita
- Division of Neurotoxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), 3900 NCTR Road Jefferson, AR 72079-9501, USA
| | - Gregory T Collins
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., Mail Code 7764, San Antonio, TX 78229, USA; South Texas Veterans Health Care System, 7400 Merton Minter Dr, San Antonio, TX 78229, USA
| |
Collapse
|
44
|
Hiranita T. Medications Discovery: Importance of Assessment of Drug Self Administration Dose-Effect Curves. ACTA ACUST UNITED AC 2015; 3. [PMID: 27413771 PMCID: PMC4940040 DOI: 10.4172/2329-6488.1000e121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Takato Hiranita
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079-9501, USA
| |
Collapse
|
45
|
Reith ME, Blough BE, Hong WC, Jones KT, Schmitt KC, Baumann MH, Partilla JS, Rothman RB, Katz JL. Behavioral, biological, and chemical perspectives on atypical agents targeting the dopamine transporter. Drug Alcohol Depend 2015; 147:1-19. [PMID: 25548026 PMCID: PMC4297708 DOI: 10.1016/j.drugalcdep.2014.12.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/04/2014] [Accepted: 12/04/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Treatment of stimulant-use disorders remains a formidable challenge, and the dopamine transporter (DAT) remains a potential target for antagonist or agonist-like substitution therapies. METHODS This review focuses on DAT ligands, such as benztropine, GBR 12909, modafinil, and DAT substrates derived from phenethylamine or cathinone that have atypical DAT-inhibitor effects, either in vitro or in vivo. The compounds are described from a molecular mechanistic, behavioral, and medicinal-chemical perspective. RESULTS Possible mechanisms for atypicality at the molecular level can be deduced from the conformational cycle for substrate translocation. For each conformation, a crystal structure of a bacterial homolog is available, with a possible role of cholesterol, which is also present in the crystal of Drosophila DAT. Although there is a direct relationship between behavioral potencies of most DAT inhibitors and their DAT affinities, a number of compounds bind to the DAT and inhibit dopamine uptake but do not share cocaine-like effects. Such atypical behavior, depending on the compound, may be related to slow DAT association, combined sigma-receptor actions, or bias for cytosol-facing DAT. Some structures are sterically small enough to serve as DAT substrates but large enough to also inhibit transport. Such compounds may display partial DA releasing effects, and may be combined with release or uptake inhibition at other monoamine transporters. CONCLUSIONS Mechanisms of atypical DAT inhibitors may serve as targets for the development of treatments for stimulant abuse. These mechanisms are novel and their further exploration may produce compounds with unique therapeutic potential as treatments for stimulant abuse.
Collapse
Affiliation(s)
- Maarten E.A. Reith
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA,Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA,Corresponding author: Maarten E.A. Reith, Department of Psychiatry, Alexandria Center of Life Sciences, New York University School of Medicine, 450 E 29th Street, Room 803, New York, NY 10016. Tel.: 212 - 263 8267; Fax: 212 – 263 8183;
| | - Bruce E. Blough
- Center for Drug Discovery, Research Triangle Institute, Research Triangle Park, NC 27709, USA
| | - Weimin C. Hong
- Psychobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Kymry T. Jones
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA
| | - Kyle C. Schmitt
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA
| | - Michael H. Baumann
- Medicinal Chemistry Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - John S. Partilla
- Medicinal Chemistry Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Richard B. Rothman
- Medicinal Chemistry Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jonathan L. Katz
- Psychobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
46
|
Hiranita T. Preclinical Efficacy of Novel Vesicular Monoamine Transporter 2 Inhibitors as Antagonists of d-Methamphetamine Self-Administration in Rats. JOURNAL OF ALCOHOLISM AND DRUG DEPENDENCE 2015; 3. [PMID: 27398393 PMCID: PMC4934384 DOI: 10.4172/2329-6488.1000e127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Takato Hiranita
- Division of Neurotoxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), 3900 NCTR Road, Jefferson, AR 72079-9501, USA
| |
Collapse
|
47
|
Hiranita T, Thorn DA. Trace Amine-Associated Receptor Type 1 as A Target for The Development of Treatments for Stimulant Abuse. JOURNAL OF ALCOHOLISM AND DRUG DEPENDENCE 2015; 3. [PMID: 27390752 PMCID: PMC4932863 DOI: 10.4172/2329-6488.1000e122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Takato Hiranita
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079-9501, USA
| | - David A Thorn
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079-9501, USA
| |
Collapse
|
48
|
Hiranita T. Cocaine Antagonists; Studies on Cocaine Self-Administration. JOURNAL OF ALCOHOLISM AND DRUG DEPENDENCE 2015; 3. [PMID: 27398394 DOI: 10.4172/2329-6488.1000e125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Takato Hiranita
- Division of Neurotoxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), USA
| |
Collapse
|
49
|
Hiranita T. Self-Administration of an Endogenous Cannabinoid 2-Arachidonoylglycerol in Experimentally Naïve Rats. JOURNAL OF ALCOHOLISM AND DRUG DEPENDENCE 2015; 3. [PMID: 27376099 PMCID: PMC4930111 DOI: 10.4172/2329-6488.1000e126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Takato Hiranita
- Division of Neurotoxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), 3900 NCTR Road, Jefferson, AR 72079-9501, USA
| |
Collapse
|
50
|
Kohut SJ, Hiranita T, Hong SK, Ebbs AL, Tronci V, Green J, Garcés-Ramírez L, Chun LE, Mereu M, Newman AH, Katz JL, Tanda G. Preference for distinct functional conformations of the dopamine transporter alters the relationship between subjective effects of cocaine and stimulation of mesolimbic dopamine. Biol Psychiatry 2014; 76:802-9. [PMID: 24853388 PMCID: PMC4353924 DOI: 10.1016/j.biopsych.2014.03.031] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 03/08/2014] [Accepted: 03/13/2014] [Indexed: 11/26/2022]
Abstract
BACKGROUND Subjective effects of cocaine are mediated primarily by dopamine (DA) transporter (DAT) blockade. The present study assessed the hypothesis that different DAT conformational equilibria regulate differences in cocaine-like subjective effects and extracellular DA induced by diverse DA-uptake inhibitors (DUIs). METHODS The relationship between cocaine-like subjective effects and stimulation of mesolimbic DA levels by standard DUIs (cocaine, methylphenidate, WIN35,428) and atypical DUIs (benztropine analogs: AHN1-055, AHN2-005, JHW007) was investigated using cocaine discrimination and DA microdialysis procedures in rats. RESULTS All drugs stimulated DA levels with different maxima and time courses. Standard DUIs, which preferentially bind outward-facing DAT conformations, fully substituted for cocaine, consistently producing cocaine-like subjective effects at DA levels of 100-125% over basal values, regardless of dose or pretreatment time. The atypical DUIs, with DAT binding minimally affected by DAT conformation, produced inconsistent cocaine-like subjective effects. Full effects were obtained, if at all, only at a few doses and pretreatment times and at DA levels 600-700% greater than basal values. Importantly, the linear, time-independent, relationship between cocaine-like subjective effects and DA stimulation obtained with standard DUIs was not obtained with the atypical DUIs. CONCLUSIONS These results suggest a time-related desensitization process underlying the reduced cocaine subjective effects of atypical DUIs that may be differentially induced by the binding modalities identified using molecular approaches. Since the DAT is the target of several drugs for treating neuropsychiatric disorders, such as attention-deficit/hyperactivity disorder, these results help to identify safe and effective medications with minimal cocaine-like subjective effects that contribute to abuse liability.
Collapse
Affiliation(s)
- Stephen J Kohut
- Psychobiology, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland
| | - Takato Hiranita
- Psychobiology, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland
| | - Soo-Kyung Hong
- Psychobiology, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland; Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Aaron L Ebbs
- Psychobiology, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland
| | - Valeria Tronci
- Psychobiology, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland
| | - Jennifer Green
- Psychobiology, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland
| | - Linda Garcés-Ramírez
- Psychobiology, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, México
| | - Lauren E Chun
- Psychobiology, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland
| | - Maddalena Mereu
- Psychobiology, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland
| | - Amy H Newman
- Medicinal Chemistry Sections, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland; Medications Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland
| | - Jonathan L Katz
- Psychobiology, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland
| | - Gianluigi Tanda
- Psychobiology, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland; Medications Development Program, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse/Intramural Research Program/National Institutes of Health/Department of Health and Human Services, Baltimore, Maryland.
| |
Collapse
|