1
|
Fultz EK, Nei AYT, Chi JC, Lichter JN, Szumlinski KK. Effects of systemic pretreatment with the NAALADase inhibitor 2-PMPA on oral methamphetamine reinforcement in C57BL/6J mice. Front Psychiatry 2024; 15:1297275. [PMID: 38638417 PMCID: PMC11024460 DOI: 10.3389/fpsyt.2024.1297275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/21/2024] [Indexed: 04/20/2024] Open
Abstract
Introduction Repeated exposure to methamphetamine (MA) in laboratory rodents induces a sensitization of glutamate release within the corticoaccumbens pathway that drives both the rewarding and reinforcing properties of this highly addictive drug. Such findings argue the potential for pharmaceutical agents inhibiting glutamate release or its postsynaptic actions at glutamate receptors as treatment strategies for MA use disorder. One compound that may accomplish both of these pharmacological actions is the N-acetylated-alpha-linked-acidic dipeptidase (NAALADase) inhibitor 2-(phosphonomethyl)pentanedioic acid (2-PMPA). 2-PMPA elevates brain levels of the endogenous agonist of glutamate mGluR3 autoreceptors, N-acetyl-aspartatylglutamate (NAAG), while potentially acting as an NMDA glutamate receptor antagonist. Of relevance to treating psychomotor stimulant use disorders, 2-PMPA is reported to reduce indices of both cocaine and synthetic cathinone reward, as well as cocaine reinforcement in preclinical rodent studies. Method Herein, we conducted three experiments to pilot the effects of systemic pretreatment with 2-PMPA (0-100 mg/kg, IP) on oral MA self-administration in C57BL/6J mice. The first experiment employed female mice with a prolonged history of MA exposure, while the mice in the second (females) and third (males and females) experiment were MA-naïve prior to study. In all experiments, mice were trained daily to nose-poke for delivery of unadulterated MA solutions until responding stabilized. Then, mice were pretreated with 2-PMPA prior to operant-conditioning sessions in which nose-poking behavior was reinforced by delivery of 120 mg/L or 200 mg/L MA (respectively, in Experiments 1 and 2/3). Results Contrary to our expectations, 30 mg/kg 2-PMPA pretreatment altered neither appetitive nor consummatory measures related to MA self-administration. In Experiment 3, 100 mg/kg 2-PMPA reduced responding in the MA-reinforced hole, as well as the number of reinforcers earned, but did not significantly lower drug intake. Discussion These results provide mixed evidenced related to the efficacy of this NAALADase inhibitor for reducing oral MA reinforcement in female mice.
Collapse
Affiliation(s)
- Elissa K. Fultz
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Andrea Y. T. Nei
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Joyce C. Chi
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Jacqueline N. Lichter
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Karen K. Szumlinski
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, United States
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
2
|
Peng Y, Wang Y, Liu L, Yang G, Zhang D, Hong S, Li L. The effects of ketamine and methamphetamine on neurotransmitters, glutamate receptors, and conditioned place preference in rat. Leg Med (Tokyo) 2023; 65:102328. [PMID: 37778205 DOI: 10.1016/j.legalmed.2023.102328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/10/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Combined methamphetamine (MA) and ketamine (KET) abuse is a serious issue. At present, however, few studies have explored the mechanism underlying their combined addiction. We established a rat conditioned place preference (CPP) model. We investigated the role of dopamine (DA), 5-hydroxytryptamine (5-HT), monoamine oxidase (MAO), glutamate receptor 1 (GluR1), and glutamate receptor 2 (GluR2) in combined MA and KET addiction. The expression levels of DA, 5-HT, and MAO were detected by enzyme-linked immunosorbent assay (ELISA), and the expressions levels of GluR1 and GluR2 were detected by western blotting. Our results showed that MA and KET successfully induced CPP in rats respectively, and KET enhanced MA-induced CPP effects, although not significantly, and KET can reduce the MA-induced increase in DA, 5-HT, MAO and promoted the MA-induced increase in GluR1 and GluR2. Therefore, it suggested that DA, 5-HT, MAO, GluR1, and GluR2 expression may be involved in the mechanism underlying MA and KET-induced drug addiction in rats. Moreover, When MA and KET are used in combination, KET appears to play a dual addictive and anti-addictive role in the regulation of MA addiction.
Collapse
Affiliation(s)
- Yanxia Peng
- School of Forensic Medicine/NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Yihang Wang
- School of Forensic Medicine/NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China; School of Basic Medicine, Hubei University of Arts and Science, Hubei, China
| | - Linlin Liu
- School of Forensic Medicine/NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Genmeng Yang
- School of Forensic Medicine/NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Dongxian Zhang
- School of Forensic Medicine/NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Shijun Hong
- School of Forensic Medicine/NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China.
| | - Lihua Li
- School of Forensic Medicine/NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China.
| |
Collapse
|
3
|
Pla-Tenorio J, Roig AM, García-Cesaní PA, Santiago LA, Sepulveda-Orengo MT, Noel RJ. Astrocytes: Role in pathogenesis and effect of commonly misused drugs in the HIV infected brain. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 5:100108. [PMID: 38020814 PMCID: PMC10663134 DOI: 10.1016/j.crneur.2023.100108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 06/05/2023] [Accepted: 08/18/2023] [Indexed: 12/01/2023] Open
Abstract
The roles of astrocytes as reservoirs and producers of a subset of viral proteins in the HIV infected brain have been studied extensively as a key to understanding HIV-associated neurocognitive disorders (HAND). However, their comprehensive role in the context of intersecting substance use and neurocircuitry of the reward pathway and HAND has yet to be fully explained. Use of methamphetamines, cocaine, or opioids in the context of HIV infection have been shown to lead to a faster progression of HAND. Glutamatergic, dopaminergic, and GABAergic systems are implicated in the development of HAND-induced cognitive impairments. A thorough review of scientific literature exploring the variety of mechanisms in which these drugs exert their effects on the HIV brain and astrocytes has revealed marked areas of convergence in overexcitation leading to increased drug-seeking behavior, inflammation, apoptosis, and irreversible neurotoxicity. The present review investigates astrocytes, the neural pathways, and mechanisms of drug disruption that ultimately play a larger holistic role in terms of HIV progression and drug use. There are opportunities for future research, therapeutic intervention, and preventive strategies to diminish HAND in the subset population of patients with HIV and substance use disorder.
Collapse
Affiliation(s)
- Jessalyn Pla-Tenorio
- Ponce Health Sciences University, School of Medicine, Department of Basic Sciences, 395 Industrial Reparada, Zona 2, Ponce, PR, 00716, Puerto Rico
| | - Angela M. Roig
- Seattle Children's Hospital, MS OC.7.830, 4800 Sand Point Way NE, Seattle, WA, 98105-0371, United States
| | - Paulina A. García-Cesaní
- Bella Vista Hospital, Family Medicine Residency, Carr. 349 Km 2.7, Cerro Las Mesas, Mayaguez, PR, 00681, Puerto Rico
| | - Luis A. Santiago
- Ponce Health Sciences University, School of Medicine, Department of Basic Sciences, 395 Industrial Reparada, Zona 2, Ponce, PR, 00716, Puerto Rico
| | - Marian T. Sepulveda-Orengo
- Ponce Health Sciences University, School of Medicine, Department of Basic Sciences, 395 Industrial Reparada, Zona 2, Ponce, PR, 00716, Puerto Rico
| | - Richard J. Noel
- Ponce Health Sciences University, School of Medicine, Department of Basic Sciences, 395 Industrial Reparada, Zona 2, Ponce, PR, 00716, Puerto Rico
| |
Collapse
|
4
|
Fontana IC, Souza DG, Souza DO, Gee A, Zimmer ER, Bongarzone S. A Medicinal Chemistry Perspective on Excitatory Amino Acid Transporter 2 Dysfunction in Neurodegenerative Diseases. J Med Chem 2023; 66:2330-2346. [PMID: 36787643 PMCID: PMC9969404 DOI: 10.1021/acs.jmedchem.2c01572] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The excitatory amino acid transporter 2 (EAAT2) plays a key role in the clearance and recycling of glutamate - the major excitatory neurotransmitter in the mammalian brain. EAAT2 loss/dysfunction triggers a cascade of neurodegenerative events, comprising glutamatergic excitotoxicity and neuronal death. Nevertheless, our current knowledge regarding EAAT2 in neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD), is restricted to post-mortem analysis of brain tissue and experimental models. Thus, detecting EAAT2 in the living human brain might be crucial to improve diagnosis/therapy for ALS and AD. This perspective article describes the role of EAAT2 in physio/pathological processes and provides a structure-activity relationship of EAAT2-binders, bringing two perspectives: therapy (activators) and diagnosis (molecular imaging tools).
Collapse
Affiliation(s)
- Igor C Fontana
- School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom.,Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil.,Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16 - Neo floor seventh, 141 83 Stockholm, Sweden
| | - Débora G Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil.,Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Av. Ipiranga, 6681 Porto Alegre, Brazil
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil
| | - Antony Gee
- School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Eduardo R Zimmer
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600, 90035-003 Porto Alegre, Brazil.,Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Av. Sarmento Leite 500, sala, 90035-003 Porto Alegre, Brazil.,Graduate Program in Biological Sciences: Biochemistry (PPGBioq), and Pharmacology and Therapeutics (PPGFT), Universidade Federal do Rio Grande do Sul, Av. Sarmento Leite 500, sala, 305 Porto Alegre, Brazil.,Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Av. Ipiranga, 6681 Porto Alegre, Brazil.,McGill University Research Centre for Studies in Aging, McGill University, Montreal, Quebec H4H 1R3, Canada
| | - Salvatore Bongarzone
- School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| |
Collapse
|
5
|
Wiah S, Roper A, Zhao P, Shekarabi A, Watson MN, Farkas DJ, Potula R, Reitz AB, Rawls SM. Troriluzole inhibits methamphetamine place preference in rats and normalizes methamphetamine-evoked glutamate carboxypeptidase II (GCPII) protein levels in the mesolimbic pathway. Drug Alcohol Depend 2023; 242:109719. [PMID: 36521236 PMCID: PMC9850846 DOI: 10.1016/j.drugalcdep.2022.109719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/04/2022] [Accepted: 11/27/2022] [Indexed: 12/12/2022]
Abstract
Riluzole, approved to manage amyotrophic lateral sclerosis, is mechanistically unique among glutamate-based therapeutics because it reduces glutamate transmission through a dual mechanism (i.e., reduces glutamate release and enhances glutamate reuptake). The profile of riluzole is favorable for normalizing glutamatergic dysregulation that perpetuates methamphetamine (METH) dependence, but pharmacokinetic and metabolic liabilities hinder repurposing. To mitigate these limitations, we synthesized troriluzole (TRLZ), a third-generation prodrug of riluzole, and tested the hypothesis that TRLZ inhibits METH hyperlocomotion and conditioned place preference (CPP) and normalizes METH-induced changes in mesolimbic glutamate biomarkers. TRLZ (8, 16 mg/kg) reduced hyperlocomotion caused by METH (1 mg/kg) without affecting spontaneous activity. TRLZ (1, 4, 8, 16 mg/kg) administered during METH conditioning (0.5 mg/kg x 4 d) inhibited development of METH place preference, and TRLZ (16 mg/kg) administered after METH conditioning reduced expression of CPP. In rats with established METH place preference, TRLZ (16 mg/kg) accelerated extinction of CPP. In cellular studies, chronic METH enhanced mRNA levels of glutamate carboxypeptidase II (GCPII) in the ventral tegmental area (VTA) and prefrontal cortex (PFC). Repeated METH also caused enhancement of GCPII protein levels in the VTA that was prevented by TRLZ (16 mg/kg). TRLZ (16 mg/kg) administered during chronic METH did not affect brain or plasma levels of METH. These results indicate that TRLZ, already in clinical trials for cerebellar ataxia, reduces development, expression and maintenance of METH CPP. Moreover, normalization of METH-induced GCPII levels in mesolimbic substrates by TRLZ points toward studying GCPII as a therapeutic target of TRLZ.
Collapse
Affiliation(s)
- Sonita Wiah
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Abigail Roper
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Psychology, College of Liberal Arts, University of Massachusetts-Boston, Boston, MA, USA
| | - Pingwei Zhao
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Aryan Shekarabi
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Mia N Watson
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Daniel J Farkas
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Raghava Potula
- Department of Pathology and Laboratory Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Allen B Reitz
- Fox Chase Chemical Diversity Center, Rockville, MD, USA
| | - Scott M Rawls
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Philogene-Khalid HL, Morrison MF, Darbinian N, Selzer ME, Schroeder J, Rawls SM. The GLT-1 enhancer clavulanic acid suppresses cocaine place preference behavior and reduces GCPII activity and protein levels in the rat nucleus accumbens. Drug Alcohol Depend 2022; 232:109306. [PMID: 35051699 PMCID: PMC8885893 DOI: 10.1016/j.drugalcdep.2022.109306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/21/2021] [Accepted: 01/10/2022] [Indexed: 12/18/2022]
Abstract
The β-lactam antibiotic ceftriaxone (CTX) is a glutamate transporter subtype 1 (GLT-1) enhancer that reduces cocaine reinforcing efficacy and relapse in rats, but pharmacokinetic liabilities limit translational utility. An attractive alternative is clavulanic acid (CLAV), a structurally related β-lactamase inhibitor and component of FDA-approved Augmentin. CLAV retains the GLT-1 enhancing effects of CTX but displays greater oral bioavailability, brain penetrability and negligible antibacterial activity. CLAV reduces morphine conditioned place preference (CPP) and ethanol consumption in rats, but knowledge about the efficacy of CLAV in preclinical models of drug addiction remains sparse. Here, we investigated effects of CLAV (10 mg/kg, IP) on the acquisition, expression, and maintenance of cocaine CPP in rats, and on two glutamate biomarkers associated with cocaine dependence, GLT-1 and glutamate carboxypeptidase II (GCPII). CLAV administered during cocaine conditioning (10 mg/kg, IP x 4 d) did not affect the development of cocaine CPP. However, a single CLAV injection, administered after the conditioning phase, reduced the expression of cocaine CPP. In rats with established cocaine preference, repeated CLAV administration facilitated extinction of cocaine CPP. In the nucleus accumbens, acute CLAV exposure reduced GCPII protein levels and activity, and a 10-d CLAV treatment regimen enhanced GLT-1 levels. These results suggest that CLAV reduces expression and maintenance of cocaine CPP but lacks effect against development of CPP. Moreover, the ability of a single injection of CLAV to reduce both GCPII activity and protein levels, as well as expression of cocaine CPP, points toward studying GCPII as a therapeutic target of CLAV.
Collapse
Affiliation(s)
- Helene L. Philogene-Khalid
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Corresponding author at: Department of Psychiatry, Medical Arts Building Suite 305, Temple University, 100 East Lehigh Ave., Philadelphia, PA 19125-1012, United States.
| | - Mary F. Morrison
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Department of Psychiatry, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Nune Darbinian
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Department of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Michael E. Selzer
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Department of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Joseph Schroeder
- Behavioral Neuroscience Program, Connecticut College, New London, CT, USA
| | - Scott M. Rawls
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
7
|
Cisneros IE, Cunningham KA. Covid-19 interface with drug misuse and substance use disorders. Neuropharmacology 2021; 198:108766. [PMID: 34454912 PMCID: PMC8388132 DOI: 10.1016/j.neuropharm.2021.108766] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/09/2021] [Accepted: 08/23/2021] [Indexed: 12/26/2022]
Abstract
The coronavirus disease 2019 (Covid-19) pandemic intensified the already catastrophic drug overdose and substance use disorder (SUD) epidemic, signaling a syndemic as social isolation, economic and mental health distress, and disrupted treatment services disproportionally impacted this vulnerable population. Along with these social and societal factors, biological factors triggered by intense stress intertwined with incumbent overactivity of the immune system and the resulting inflammatory outcomes may impact the functional status of the central nervous system (CNS). We review the literature concerning SARS-CoV2 infiltration and infection in the CNS and the prospects of synergy between stress, inflammation, and kynurenine pathway function during illness and recovery from Covid-19. Taken together, inflammation and neuroimmune signaling, a consequence of Covid-19 infection, may dysregulate critical pathways and underlie maladaptive changes in the CNS, to exacerbate the development of neuropsychiatric symptoms and in the vulnerability to develop SUD. This article is part of the special Issue on 'Vulnerabilities to Substance Abuse'.
Collapse
Affiliation(s)
- I E Cisneros
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA; Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA.
| | - K A Cunningham
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
8
|
Potential Benefits of N-Acetylcysteine in Preventing Pregabalin-Induced Seeking-Like Behavior. Healthcare (Basel) 2021; 9:healthcare9040376. [PMID: 33805329 PMCID: PMC8066267 DOI: 10.3390/healthcare9040376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 11/24/2022] Open
Abstract
Substance-use disorder is globally prevalent and responsible for numerous social and medical problems. Pregabalin (Lyrica), typically used to treat diabetic neuropathy, has recently emerged as a drug of abuse. Drug abuse is associated with several neuronal changes, including the downregulation of glutamate transporters such as glutamate transporter 1 and cystine/glutamate antiporter. We investigated the effects of N-acetylcysteine, a glutamate transporter 1 and xCT upregulator, on pregabalin addiction using a conditioned place preference paradigm. Pregabalin (60 mg/kg) was found to induce conditioned place preference when compared to a vehicle. A 100 mg/kg dose of N-acetylcysteine was found to block pregabalin-seeking behaviors. These results support previous findings showing that glutamate transporters play an important role in pregabalin-induced seeking behaviors. N-acetylcysteine may represent a beneficial agent in preventing the abuse potential of pregabalin.
Collapse
|
9
|
Heinsbroek JA, De Vries TJ, Peters J. Glutamatergic Systems and Memory Mechanisms Underlying Opioid Addiction. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a039602. [PMID: 32341068 DOI: 10.1101/cshperspect.a039602] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Glutamate is the main excitatory neurotransmitter in the brain and is of critical importance for the synaptic and circuit mechanisms that underlie opioid addiction. Opioid memories formed over the course of repeated drug use and withdrawal can become powerful stimuli that trigger craving and relapse, and glutamatergic neurotransmission is essential for the formation and maintenance of these memories. In this review, we discuss the mechanisms by which glutamate, dopamine, and opioid signaling interact to mediate the primary rewarding effects of opioids, and cover the glutamatergic systems and circuits that mediate the expression, extinction, and reinstatement of opioid seeking over the course of opioid addiction.
Collapse
Affiliation(s)
- Jasper A Heinsbroek
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Taco J De Vries
- Amsterdam Neuroscience, Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Faculty of Earth and Life Sciences, VU University, 1081HV Amsterdam, The Netherlands.,Amsterdam Neuroscience, Department of Anatomy and Neurosciences, VU University Medical Center, 1081HZ Amsterdam, The Netherlands
| | - Jamie Peters
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| |
Collapse
|
10
|
Abstract
The pervasive and devastating nature of substance use disorders underlies the need for the continued development of novel pharmacotherapies. We now know that glia play a much greater role in neuronal processes than once believed. The various types of glial cells (e.g., astrocytes, microglial, oligodendrocytes) participate in numerous functions that are crucial to healthy central nervous system function. Drugs of abuse have been shown to interact with glia in ways that directly contribute to the pharmacodynamic effects responsible for their abuse potential. Through their effect upon glia, drugs of abuse also alter brain function resulting in behavioral changes associated with substance use disorders. Therefore, drug-induced changes in glia and inflammation within the central nervous system (neuroinflammation) have been investigated to treat various aspects of drug abuse and dependence. This article presents a brief overview of the effects of each of the major classes of addictive drugs on glia. Next, the paper reviews the pre-clinical and clinical studies assessing the effects that glial modulators have on abuse-related behavioral effects, such as pleasure, withdrawal, and motivation. There is a strong body of pre-clinical literature demonstrating the general effectiveness of several glia-modulating drugs in models of reward and relapse. Clinical studies have also yielded promising results, though not as robust. There is still much to disentangle regarding the integration between addictive drugs and glial cells. Improved understanding of the relationship between glia and the pathophysiology of drug abuse should allow for more precise exploration in the development and testing of glial-directed treatments for substance use disorders.
Collapse
Affiliation(s)
- Jermaine D. Jones
- Division on Substance Use Disorders, New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|
11
|
Yang L, Guo N, Fan W, Ni C, Huang M, Bai L, Zhang L, Zhang X, Wen Y, Li Y, Zhou X, Bai J. Thioredoxin-1 blocks methamphetamine-induced injury in brain through inhibiting endoplasmic reticulum and mitochondria-mediated apoptosis in mice. Neurotoxicology 2020; 78:163-169. [PMID: 32203791 DOI: 10.1016/j.neuro.2020.03.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/12/2020] [Accepted: 03/12/2020] [Indexed: 02/06/2023]
Abstract
Methamphetamine (METH) has been reported to induce endoplasmic reticulum (ER) stress and neuronal apoptosis in the central nervous system (CNS) during the development of addiction. Thioredoxin-1 (Trx-1) is a redox regulating protein and plays an important role in inhibiting apoptosis and protects neurons from cytotoxicity through ER and mitochondria-mediated pathways. Our previous study has been reported that Trx-1 protects mice from METH-induced rewarding effect. However, whether Trx-1 plays the role in resisting METH injury is still unclear. Here, we aim to investigate whether Trx-1 participates in the regulation of METH-induced CNS injury via ER stress and mitochondria-mediated pathways. Our study first repeated the conditioned place preference expression induced by METH. Then we detected and found that METH increased the expression of N-methyl-d-asparate (NMDA) receptor subunit 2B (NR2B) and the level of glutamate (Glu) in the ventral tegmental area (VTA) and nucleus accumbens (NAc), while Trx-1 overexpression suppressed the increases. We further examined ER stress-related proteins and mitochondrial apoptosis pathway in the VTA and NAc, and found that METH increased the expressions of glucose regulated protein 78 (GRP78), C/EBP homologous protein (CHOP), and Bax, as same time decreased the expressions of procaspase12, Bcl-2, and procaspase3, while Trx-1 overexpression blocked these changes. These results indicate that Trx-1 blocks METH-induced injury by suppressing ER stress and mitochondria-mediated apoptosis in the VTA and NAc via targeting glutamatergic system.
Collapse
Affiliation(s)
- Lihua Yang
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China; Narcotics Control School, Yunnan Police College, Kunming, 650223, China
| | - Ningning Guo
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Wei Fan
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Chunmin Ni
- Narcotics Control School, Yunnan Police College, Kunming, 650223, China
| | - Mengbing Huang
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Liping Bai
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Le Zhang
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Xianwen Zhang
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Yunbo Wen
- Narcotics Control School, Yunnan Police College, Kunming, 650223, China
| | - Ye Li
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Xiaoshuang Zhou
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
12
|
Althobaiti YS, Almalki AH. Effects of environmental enrichment on reinstatement of methamphetamine-induced conditioned place preference. Behav Brain Res 2020; 379:112372. [PMID: 31759048 DOI: 10.1016/j.bbr.2019.112372] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/15/2019] [Accepted: 11/19/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The influence of environmental enrichment (EE) on reinstatement to methamphetamine (METH) seeking in rat model was investigated. METHODS Wistar rats were divided to receive saline (1 ml/kg) or METH (1 mg/kg, i.p.) for 8 days during the conditioning training in the conditioned place preference (CPP) paradigm, which is one of the most popular models to study the motivational effects of drugs and non-drug treatments in experimental animals. Rats were then kept in either isolated (IE) or enriched environment (EE) for 30 days during the extinction training. Animals were finally examined for reinstatement provoked by i.p. injections of METH. RESULTS Saline injections during the conditioning phase did not change CPP during reinstatement in animals of IE or EE control groups. METH injections reinstated place preference in the IE animal group. Interestingly, EE significantly blocked this reinstatement effects of METH. CONCLUSION These results show the important role of social interactions and positive environment conditions in preventing reinstatement to drug use.
Collapse
Affiliation(s)
- Yusuf S Althobaiti
- Taif University, College of Pharmacy, Department of Pharmacology and Toxicology, Taif, Saudi Arabia; Taif University, College of Pharmacy, Addiction and Neuroscience Research Unit, Taif, Saudi Arabia.
| | - Atiah H Almalki
- Taif University, College of Pharmacy, Addiction and Neuroscience Research Unit, Taif, Saudi Arabia; Taif University, College of Pharmacy, Department of Pharmaceutical chemistry, Taif, Saudi Arabia
| |
Collapse
|
13
|
Brown CN, Fultz EK, Ferdousian S, Rogers S, Lustig E, Page A, Shahin JR, Flaherty DM, Von Jonquieres G, Bryant CD, Kippin TE, Szumlinski KK. Transgenic Analyses of Homer2 Function Within Nucleus Accumbens Subregions in the Regulation of Methamphetamine Reward and Reinforcement in Mice. Front Psychiatry 2020; 11:11. [PMID: 32116834 PMCID: PMC7013000 DOI: 10.3389/fpsyt.2020.00011] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/07/2020] [Indexed: 01/07/2023] Open
Abstract
Problems associated with the abuse of amphetamine-type stimulants, including methamphetamine (MA), pose serious health and socioeconomic issues world-wide. While it is well-established that MA's psychopharmacological effects involve interactions with monoamine neurotransmission, accumulating evidence from animal models implicates dysregulated glutamate in MA addiction vulnerability and use disorder. Recently, we discovered an association between genetic vulnerability to MA-taking and increased expression of the glutamate receptor scaffolding protein Homer2 within both the shell and core subregions of the nucleus accumbens (NAC) and demonstrated a necessary role for Homer2 within the shell subregion in MA reward and reinforcement in mice. This report extends our earlier work by interrogating the functional relevance of Homer2 within the NAC core for the conditioned rewarding and reinforcing properties of MA. C57BL/6J mice with a virus-mediated knockdown of Homer2b expression within the NAC core were first tested for the development and expression of a MA-induced conditioned place-preference/CPP (four pairings of 2 mg/kg MA) and then were trained to self-administer oral MA under operant-conditioning procedures (5-80 mg/L). Homer2b knockdown in the NAC core augmented a MA-CPP and shifted the dose-response function for MA-reinforced responding, above control levels. To determine whether Homer2b within NAC subregions played an active role in regulating MA reward and reinforcement, we characterized the MA phenotype of constitutive Homer2 knockout (KO) mice and then assayed the effects of virus-mediated overexpression of Homer2b within the NAC shell and core of wild-type and KO mice. In line with the results of NAC core knockdown, Homer2 deletion potentiated MA-induced CPP, MA-reinforced responding and intake, as well as both cue- and MA-primed reinstatement of MA-seeking following extinction. However, there was no effect of Homer2b overexpression within the NAC core or the shell on the KO phenotype. These data provide new evidence indicating a globally suppressive role for Homer2 in MA-seeking and MA-taking but argue against specific NAC subregions as the neural loci through which Homer2 actively regulates MA addiction-related behaviors.
Collapse
Affiliation(s)
- Chelsea N Brown
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Elissa K Fultz
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Sami Ferdousian
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Sarina Rogers
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Elijah Lustig
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Ariana Page
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - John R Shahin
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Daniel M Flaherty
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Georg Von Jonquieres
- Translational Neuroscience Facility, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Camron D Bryant
- Laboratory of Addiction Genetics, Departments of Pharmacology and Experimental Therapeutics and Psychiatry, Boston University School of Medicine, Boston, MA, United States
| | - Tod E Kippin
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States.,Department of Molecular, Cellular and Developmental Biology and the Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States.,Center for Collaborative Biotechnology, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Karen K Szumlinski
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States.,Department of Molecular, Cellular and Developmental Biology and the Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
14
|
Arezoomandan R, Aliaghaei A, Khodagholi F, Haghparast A. Minocycline induces the expression of intra-accumbal glutamate transporter-1 in the morphine-dependent rats. Asian J Psychiatr 2019; 46:70-73. [PMID: 31630007 DOI: 10.1016/j.ajp.2019.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/06/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023]
Abstract
Glial glutamate transporters (GLT-1) is responsible for glutamate homeostasis. GLT-1 expression and glutamate uptake can be affected by addictive drugs and can be used as a target in addiction pharmacotherapy. It has been shown that minocycline, an antibiotic with anti-inflammatory, and neuroprotective properties, can upregulate the expression of GLT-1. In the present study, in morphine-dependent rats, the effect of minocycline on expression of GLT-1 in nucleus accumbens was investigated by immunohistochemistry. The expression of GLT-1 significantly increased in minocycline treated animals. In line with other studies, our findings showed that restoring GLT-1 expression with minocycline might be considered as a potential target for correcting pre-clinical and clinical manifestations of drug addiction.
Collapse
Affiliation(s)
- Reza Arezoomandan
- Addiction Department, School of Behavioral Sciences and Mental Health (Tehran Institute of Psychiatry), Iran University of Medical Sciences, Tehran, Iran; Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Aliaghaei
- Neuroscience Lab, Biology and Anatomical Sciences Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Magrone T, Jirillo E. Drugs of Abuse Induced-Subversion of the Peripheral Immune Response and Central Glial Activity: Focus on Novel Therapeutic Approaches. Endocr Metab Immune Disord Drug Targets 2019; 19:281-291. [PMID: 30488804 DOI: 10.2174/1871530319666181129104329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Drugs of abuse affect both central nervous system (CNS) and peripheral immune function. Besides the involvement of dopamine and glutamate systems, chronic exposure to drugs of abuse alters immune homeostasis, promoting a pro-inflammatory status. At the same time, impaired peripheral immunity leads to an increased susceptibility to infections in drug abusers. DISCUSSION There is evidence that certain drugs, such as opioids, activate microglial cells and astrocytes which, in turn, provoke central neuroinflammation. Particularly, opioids bind the Toll-like receptor (TLR)-4 with increased expression of nuclear factor kappa-light-chain-enhancer of activated B cells and release of pro-inflammatory cytokines. Peripheral mediators released by immune cells also contribute to aggravate central neuroinflammation. CONCLUSION These are based either on the inhibition of TLR-4 activation by drugs of abuse or on the correction of dopamine and glutamate pathways. Finally, a hypothetic nutraceutical intervention with polyphenols in view of their anti-inflammatory and anti-oxidant properties will be outlined as an adjuvant treatment for drugs of abuse-related disorders.
Collapse
Affiliation(s)
- Thea Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, School of Medicine, Bari, Italy
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, School of Medicine, Bari, Italy
| |
Collapse
|
16
|
Althobaiti YS. Role of venlafaxine in relapse to methamphetamine seeking. Potential treatment option for drug dependence. Saudi Med J 2019; 40:339-346. [PMID: 30957126 PMCID: PMC6506650 DOI: 10.15537/smj.2019.4.23718] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES To investigate the effects of venlafaxine (VEN) on the relapse of methamphetamine (METH)-induced conditioned place preference (CPP) in rats. METHODS This study was conducted at the Department of Pharmacology and Toxicology, and the Addiction and Neuroscience Research Unit, College of Pharmacy, Taif University, Taif, Kingdom of Saudi Arabia between May 2017 and October 2018. A total of 32 male Wistar rats were used in this study. Rats were divided to receive either METH or saline during the conditioning phase. Following the acquisition of METH-induced CPP, the animals were randomly divided to receive saline or VEN for 21 days instead of METH during the extinction training. The reinstatement was initiated by re-exposure to a single dose of METH to test any anti-reinstatement effects of VEN. Results: Two-way repeated measures Analysis of Variance (ANOVA) (time × chamber) was used to analyze time spent in conditioning chambers, while distance traveled was analyzed using one-way ANOVA. Saline or VEN, when administered alone, did not affect CPP or locomotor activity results. A priming intraperitoneal injection of METH reinstated CPP in the animals treated with saline during the extinction phase. Interestingly, VEN treatment blocked METH-induced CPP. CONCLUSION Venlafaxine effects on the reinstatement of METH-induced CPP are not likely due to nonspecific effects on locomotor activity. This beneficial effect of VEN on relapse to METH-induced CPP could be due to its antidepressant effects. Venlafaxine can thus be a potential therapeutic option in the treatment of relapse to METH-seeking behaviors.
Collapse
Affiliation(s)
- Yusuf S Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, Taif, Kingdom of Saudi Arabia. E-mail.
| |
Collapse
|
17
|
Corkrum M, Rothwell PE, Thomas MJ, Kofuji P, Araque A. Opioid-Mediated Astrocyte-Neuron Signaling in the Nucleus Accumbens. Cells 2019; 8:cells8060586. [PMID: 31207909 PMCID: PMC6628279 DOI: 10.3390/cells8060586] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/08/2019] [Accepted: 06/11/2019] [Indexed: 12/29/2022] Open
Abstract
Major hallmarks of astrocyte physiology are the elevation of intracellular calcium in response to neurotransmitters and the release of neuroactive substances (gliotransmitters) that modulate neuronal activity. While μ-opioid receptor expression has been identified in astrocytes of the nucleus accumbens, the functional consequences on astrocyte–neuron communication remains largely unknown. The present study has investigated the astrocyte responsiveness to μ-opioid signaling and the regulation of gliotransmission in the nucleus accumbens. Through the combination of calcium imaging and whole-cell patch clamp electrophysiology in brain slices, we have found that μ-opioid receptor activation in astrocytes elevates astrocyte cytoplasmic calcium and stimulates the release of the gliotransmitter glutamate, which evokes slow inward currents through the activation of neuronal N-methyl-D-aspartate (NMDA) receptors. These results indicate the existence of molecular mechanisms underlying opioid-mediated astrocyte–neuron signaling in the nucleus accumbens.
Collapse
Affiliation(s)
- Michelle Corkrum
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Patrick E Rothwell
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Mark J Thomas
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Paulo Kofuji
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
18
|
Hakami AY, Alshehri FS, Sari Y. β-lactams modulate astroglial glutamate transporters and attenuate dependence to CP 55,940, a CB1 receptor agonist, in rat model. Behav Brain Res 2019; 359:709-718. [PMID: 30257184 DOI: 10.1016/j.bbr.2018.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 12/19/2022]
Abstract
Studies on cannabinoids have reported contradictory findings, showing both aversion and rewarding outcomes in conditioned place preference (CPP). Various possibilities have been suggested to explain the aversive properties of cannabinoids, including the pharmacokinetics profile and dose selection. In this study, we have established a CPP method to investigate the effects of modulating astroglial glutamate transporters in cannabinoid dependence using a cannabinoid receptor 1 (CB1R) agonist, CP 55,940 (CP). Previous reports using CPP paradigm demonstrated the involvement of glutamatergic system in seeking behavior of several drugs of abuse such as cocaine, heroin and nicotine. Glutamate homeostasis is maintained by several astroglial glutamate transporters, such as glutamate transporter 1 (GLT-1), cystine/glutamate transporter (xCT) and glutamate aspartate transporter (GLAST). In this study, we investigated the effects of Ampicillin/Sulbactam, β-lactam compounds known to upregulate GLT-1 and xCT, on cannabinoid seeking behavior using CP. We found first that one prime dose of CP induced CP reinstatement; this effect was associated, in part, with significant downregulation of xCT expression in the nucleus accumbens, dorsomedial prefrontal cortex and amygdala. Moreover, GLT-1 expression was downregulated in the amygdala. Importantly, Ampicillin/Sulbactam treatment during the extinction phase attenuated CP-induced reinstatement and restored the expression of GLT-1 and xCT in mesocorticolimbic brain regions. These findings suggest that β-lactams may play a potential therapeutic role in attenuating dependence to cannabinoids, in part, through upregulation of GLT-1 and xCT.
Collapse
Affiliation(s)
- Alqassem Y Hakami
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Fahad S Alshehri
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
19
|
Wu Q, Qi C, Long J, Liao Y, Wang X, Xie A, Liu J, Hao W, Tang Y, Yang B, Liu T, Tang J. Metabolites Alterations in the Medial Prefrontal Cortex of Methamphetamine Users in Abstinence: A 1H MRS Study. Front Psychiatry 2018; 9:478. [PMID: 30420814 PMCID: PMC6215956 DOI: 10.3389/fpsyt.2018.00478] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 09/12/2018] [Indexed: 11/13/2022] Open
Abstract
Background: The medial prefrontal cortex (mPFC) contains various neurotransmitter systems and plays an important role in drug use. Broad body of literature on how methamphetamine (MA) affects the structure and metabolism in the animal's mPFC is emerging, while the effects on metabolites of mPFC among human is still unclear. In this study, proton magnetic resonance spectroscopy (1H MRS) was used to measure metabolites of mPFC in methamphetamine dependent subjects. Methods: Sixty-one subjects with a history of MA dependence (fulfiled the Diagnostic and Statistical Manual of Mental Disorders, fourth edition criteria) and 65 drug-naïve control subjects (age19-45) completed 1H MRS scans using 3.0T Siemens MRI scanner. Single voxel spectra were acquired from the mPFC bilaterally using a point resolved spectroscopy sequence (PRESS). The 1H MRS data were automatically fit with linear combination model for quantification of metabolite levels of n-acetyl-aspartate (NAA), myo-inositol (mI), glycerophosphocholine plus phosphocholine(GPC+PC), phosphocreatine plus creatine (PCr+Cr), and glutamate (Glu). Metabolite levels were reported as ratios to PCr+Cr. Results: The MA group showed a significant reduction in NAA/PCr+Cr ratio and elevation in Glu/PCr+Cr ratio and mI/PCr+Cr ratio, compared with healthy control. No significant correlation was found between metabolite ratios and MA use variables. Conclusions: MA use is associated with a significant increased Glu/PCr+Cr ratio, mI/PCr+Cr ratio and reduced NAA/PCr+Cr ratio in the mPFC of MA dependence subjects. These findings suggest that Glu may play a key role in MA induced neurotoxicity.
Collapse
Affiliation(s)
- Qiuxia Wu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China
- Chinese National Clinical Research Center on Mental Disorders, Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center on Mental Disorders, Changsha, China
- National Technology Institute on Mental Disorders, Changsha, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
- Department of Psychological Sciences, Texas Tech University, Lubbock, TX, United States
| | - Chang Qi
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China
- Chinese National Clinical Research Center on Mental Disorders, Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center on Mental Disorders, Changsha, China
- National Technology Institute on Mental Disorders, Changsha, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Jiang Long
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China
- Chinese National Clinical Research Center on Mental Disorders, Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center on Mental Disorders, Changsha, China
- National Technology Institute on Mental Disorders, Changsha, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Yanhui Liao
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China
- Chinese National Clinical Research Center on Mental Disorders, Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center on Mental Disorders, Changsha, China
- National Technology Institute on Mental Disorders, Changsha, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Xuyi Wang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China
- Chinese National Clinical Research Center on Mental Disorders, Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center on Mental Disorders, Changsha, China
- National Technology Institute on Mental Disorders, Changsha, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - An Xie
- Department of Radiology, Hunan Provincial People's Hospital, Changsha, China
| | - Jianbin Liu
- Department of Radiology, Hunan Provincial People's Hospital, Changsha, China
| | - Wei Hao
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China
- Chinese National Clinical Research Center on Mental Disorders, Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center on Mental Disorders, Changsha, China
- National Technology Institute on Mental Disorders, Changsha, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Yiyuan Tang
- Department of Psychological Sciences, Texas Tech University, Lubbock, TX, United States
| | - Baozhu Yang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Tieqiao Liu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China
- Chinese National Clinical Research Center on Mental Disorders, Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center on Mental Disorders, Changsha, China
- National Technology Institute on Mental Disorders, Changsha, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Jinsong Tang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- Mental Health Institute, The Second Xiangya Hospital, Central South University, Changsha, China
- Chinese National Clinical Research Center on Mental Disorders, Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center on Mental Disorders, Changsha, China
- National Technology Institute on Mental Disorders, Changsha, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| |
Collapse
|
20
|
Bachtell RK, Jones JD, Heinzerling KG, Beardsley PM, Comer SD. Glial and neuroinflammatory targets for treating substance use disorders. Drug Alcohol Depend 2017; 180:156-170. [PMID: 28892721 PMCID: PMC5790191 DOI: 10.1016/j.drugalcdep.2017.08.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/28/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND The plenary session at the 2016 Behavior, Biology and Chemistry: Translational Research in Addiction Conference focused on glia as potential players in the development, persistence and treatment of substance use disorders. Glia partake in various functions that are important for healthy brain activity. Drugs of abuse alter glial cell activity producing several perturbations in brain function that are thought to contribute to behavioral changes associated with substance use disorders. Consequently, drug-induced changes in glia-driven processes in the brain represent potential targets for pharmacotherapeutics treating substance use disorders. METHODS Four speakers presented preclinical and clinical research illustrating the effects that glial modulators have on abuse-related behavioral effects of psychostimulants and opioids. This review highlights some of these findings and expands its focus to include other research focused on drug-induced glia abnormalities and glia-focused treatment approaches in substance use disorders. RESULTS Preclinical findings show that drugs of abuse induce neuroinflammatory signals and disrupt glutamate homeostasis through their interaction with microglia and astrocytes. Preclinical and clinical studies testing the effects of glial modulators show general effectiveness in reducing behaviors associated with substance use disorders. CONCLUSIONS The contribution of drug-induced glial activity continues to emerge as an intriguing target for substance use disorder treatments. Clinical investigations of glial modulators have yielded promising results on substance use measures and indicate that they are generally safe and well-tolerated. However, results have not been entirely positive and more questions remain for continued exploration in the development and testing of glial-directed treatments for substance use disorders.
Collapse
Affiliation(s)
- Ryan K. Bachtell
- Department of Psychology and Neuroscience, and Center for Neuroscience, UCB 345, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Jermaine D. Jones
- Division on Substance Use Disorders, New York State Psychiatric Institute and College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| | - Keith G. Heinzerling
- Department of Family Medicine and Center for Behavioral and Addiction Medicine, UCLA, Los Angeles, CA, USA
| | - Patrick M. Beardsley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N. 12th Street, Richmond, VA 23298, USA
| | - Sandra D. Comer
- Division on Substance Use Disorders, New York State Psychiatric Institute and College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|
21
|
Drugs to Alter Extracellular Concentration of Glutamate: Modulators of Glutamate Uptake Systems. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/978-1-4939-7228-9_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
22
|
Spencer S, Kalivas PW. Glutamate Transport: A New Bench to Bedside Mechanism for Treating Drug Abuse. Int J Neuropsychopharmacol 2017; 20:797-812. [PMID: 28605494 PMCID: PMC5632313 DOI: 10.1093/ijnp/pyx050] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/09/2017] [Indexed: 02/06/2023] Open
Abstract
Drug addiction has often been described as a "hijacking" of the brain circuits involved in learning and memory. Glutamate is the principal excitatory neurotransmitter in the brain, and its contribution to synaptic plasticity and learning processes is well established in animal models. Likewise, over the past 20 years the addiction field has ascribed a critical role for glutamatergic transmission in the development of addiction. Chronic drug use produces enduring neuroadaptations in corticostriatal projections that are believed to contribute to a maladaptive deficit in inhibitory control over behavior. Much of this research focuses on the role played by ionotropic glutamate receptors directly involved in long-term potentiation and depression or metabotropic receptors indirectly modulating synaptic plasticity. Importantly, the balance between glutamate release and clearance tightly regulates the patterned activation of these glutamate receptors, emphasizing an important role for glutamate transporters in maintaining extracellular glutamate levels. Five excitatory amino acid transporters participate in active glutamate reuptake. Recent evidence suggests that these glutamate transporters can be modulated by chronic drug use at a variety of levels. In this review, we synopsize the evidence and mechanisms associated with drug-induced dysregulation of glutamate transport. We then summarize the preclinical and clinical data suggesting that glutamate transporters offer an effective target for the treatment of drug addiction. In particular, we focus on the role that altered glutamate transporters have in causing drug cues and contexts to develop an intrusive quality that guides maladaptive drug seeking behaviors.
Collapse
Affiliation(s)
- Sade Spencer
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina.,Correspondence: Sade Spencer, PhD, Medical University of South Carolina, 173 Ashley Avenue, BSB, 403- MSC 510, Charleston, SC 29425 ()
| | - Peter W Kalivas
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
23
|
Sarkar A, Rakshit S, Bhattacharya SC. Interpreting the effect of confined cyclodextrin media on the FRET efficacy between Naproxen and a bio-active 3-pyrazolyl-2-pyrazoline derivative on the light of spectroscopic investigation appended by TD-DFT simulations and molecular docking analysis. J Photochem Photobiol A Chem 2017. [DOI: 10.1016/j.jphotochem.2017.04.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Masaki Y, Izumi Y, Matsumura A, Akaike A, Kume T. Protective effect of Nrf2–ARE activator isolated from green perilla leaves on dopaminergic neuronal loss in a Parkinson's disease model. Eur J Pharmacol 2017; 798:26-34. [DOI: 10.1016/j.ejphar.2017.02.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/25/2017] [Accepted: 02/03/2017] [Indexed: 01/02/2023]
|
25
|
Wan L, Bi J, Li J, Zuo Z. Glutamate transporter type 3 participates in maintaining morphine-induced conditioned place preference. Neuroscience 2017; 344:67-73. [PMID: 28049029 DOI: 10.1016/j.neuroscience.2016.12.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 01/15/2023]
Abstract
Glutamate transporters (EAAT) have been implicated in the drug addiction behavior. We determined whether EAAT type 3 (EAAT3) played a role in morphine addiction. Six- to eight-week-old EAAT3 knockout (EAAT3-/-) mice and their wild-type littermates received 3 intraperitoneal injections of 10mg/kg morphine, each on an alternative day, to induce conditioned place preference (CPP). Two days after the place preference returned to baseline, mice received 2.5mg/kg morphine to induce reinstatement. Some mice received intraperitoneal injection of 4mg/kg riluzole, an EAAT activator, 30min before morphine or saline injection. Hippocampus, medial prefrontal cortex, nucleus accumbens and ventral tegmental area were harvested for Western analysis 24h after the last dose of morphine was injected. Morphine induced CPP in wild-type and EAAT3-/- mice. Gender is not a statistically significant factor to influence this behavior. This conditioned behavior extinguished after morphine administration was stopped for 8-9days in wild-type mice, while this extinction occurred 6days after discontinuation of morphine injection in EAAT3-/- mice. A small dose of morphine similarly reinstated the conditioned behavior in the wild-type and EAAT3-/- mice. Riluzole abolished morphine-induced CPP during the initial place preference. Morphine increased EAAT3 expression in the plasma membrane of medial prefrontal cortex, nucleus accumbens and ventral tegmental area but did not affect EAAT3 expression in the hippocampus. These results suggest that EAAT3 delays the extinction of morphine-induced CPP. EAAT activation may prevent the formation of morphine-induced CPP.
Collapse
Affiliation(s)
- Li Wan
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, United States; Department of Anesthesiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiangjiang Bi
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, United States; Department of Anesthesiology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Li
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, United States
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, United States.
| |
Collapse
|
26
|
Mohammadi R, Jahanshahi M, Jameie SB. 5-HT2A Serotonin Receptor Density in Adult Male Rats' Hippocampus after Morphine-based Conditioned Place Preference. Basic Clin Neurosci 2016; 7:249-58. [PMID: 27563418 PMCID: PMC4981837 DOI: 10.15412/j.bcn.03070310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 04/01/2015] [Accepted: 08/02/2015] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION A close interaction exists between the brain opioid and serotonin (5-HT) neurotransmitter systems. Brain neurotransmitter 5-HT plays an important role in the regulation of reward-related processing. However, a few studies have investigated the potential role of 5-HT2A receptors in this behavior. Therefore, the aim of the present study was to assess the influence of morphine and Conditioned Place Preference (CPP) on the density of 5-HT2A receptor in neurons of rat hippocampal formation. METHODS Morphine (10 mg/kg, IP) was injected in male Wistar rats for 7 consecutive days (intervention group), but control rats received just normal saline (1 mL/kg, IP). We used a hotplate test of analgesia to assess induction of tolerance to analgesic effects of morphine on days 1 and 8 of injections. Later, two groups of rats were sacrificed one day after 7 days of injections, their whole brains removed, and the striatum and PFC immediately dissected. Then, the NR1 gene expression was examined with a semi-quantitative RT-PCR method. RESULTS Our data showed that the maximum response was obtained with 2.5 mg/kg of morphine. The density of 5-HT2A receptor in different areas of the hippocampus increased significantly at sham-morphine and CPP groups (P<0.05). On the other hand, the CPP groups had more 5-HT2A receptors than sham-morphine groups and also the sham-morphine groups had more 5-HT2A receptors than the control groups. CONCLUSION We concluded that the phenomenon of conditioned place preference induced by morphine can cause a significant increase in the number of serotonin 5-HT2A receptors in neurons of all areas of hippocampus.
Collapse
Affiliation(s)
- Rabie Mohammadi
- Department of Anatomy, Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Jahanshahi
- Department of Anatomy, Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Seyed Behnamedin Jameie
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Scofield MD, Heinsbroek JA, Gipson CD, Kupchik YM, Spencer S, Smith ACW, Roberts-Wolfe D, Kalivas PW. The Nucleus Accumbens: Mechanisms of Addiction across Drug Classes Reflect the Importance of Glutamate Homeostasis. Pharmacol Rev 2016; 68:816-71. [PMID: 27363441 PMCID: PMC4931870 DOI: 10.1124/pr.116.012484] [Citation(s) in RCA: 413] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The nucleus accumbens is a major input structure of the basal ganglia and integrates information from cortical and limbic structures to mediate goal-directed behaviors. Chronic exposure to several classes of drugs of abuse disrupts plasticity in this region, allowing drug-associated cues to engender a pathologic motivation for drug seeking. A number of alterations in glutamatergic transmission occur within the nucleus accumbens after withdrawal from chronic drug exposure. These drug-induced neuroadaptations serve as the molecular basis for relapse vulnerability. In this review, we focus on the role that glutamate signal transduction in the nucleus accumbens plays in addiction-related behaviors. First, we explore the nucleus accumbens, including the cell types and neuronal populations present as well as afferent and efferent connections. Next we discuss rodent models of addiction and assess the viability of these models for testing candidate pharmacotherapies for the prevention of relapse. Then we provide a review of the literature describing how synaptic plasticity in the accumbens is altered after exposure to drugs of abuse and withdrawal and also how pharmacological manipulation of glutamate systems in the accumbens can inhibit drug seeking in the laboratory setting. Finally, we examine results from clinical trials in which pharmacotherapies designed to manipulate glutamate systems have been effective in treating relapse in human patients. Further elucidation of how drugs of abuse alter glutamatergic plasticity within the accumbens will be necessary for the development of new therapeutics for the treatment of addiction across all classes of addictive substances.
Collapse
Affiliation(s)
- M D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - J A Heinsbroek
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - C D Gipson
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - Y M Kupchik
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - S Spencer
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - A C W Smith
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - D Roberts-Wolfe
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| | - P W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina (M.D.S., J.A.H., S.S., D.R.-W., P.W.K.); Department of Psychology, Arizona State University, Tempe, Arizona (C.D.G.); Department of Neuroscience, Hebrew University, Jerusalem, Israel (Y.M.K.); and Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York (A.C.W.S.)
| |
Collapse
|
28
|
Karklin Fontana AC, Fox DP, Zoubroulis A, Valente Mortensen O, Raghupathi R. Neuroprotective Effects of the Glutamate Transporter Activator (R)-(-)-5-methyl-1-nicotinoyl-2-pyrazoline (MS-153) following Traumatic Brain Injury in the Adult Rat. J Neurotrauma 2016; 33:1073-83. [PMID: 26200170 PMCID: PMC4892232 DOI: 10.1089/neu.2015.4079] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Traumatic brain injury (TBI) in humans and in animals leads to an acute and sustained increase in tissue glutamate concentrations within the brain, triggering glutamate-mediated excitotoxicity. Excitatory amino acid transporters (EAATs) are responsible for maintaining extracellular central nervous system glutamate concentrations below neurotoxic levels. Our results demonstrate that as early as 5 min and up to 2 h following brain trauma in brain-injured rats, the activity (Vmax) of EAAT2 in the cortex and the hippocampus was significantly decreased, compared with sham-injured animals. The affinity for glutamate (KM) and the expression of glutamate transporter 1 (GLT-1) and glutamate aspartate transporter (GLAST) were not altered by the injury. Administration of (R)-(-)-5-methyl-1-nicotinoyl-2-pyrazoline (MS-153), a GLT-1 activator, beginning immediately after injury and continuing for 24 h, significantly decreased neurodegeneration, loss of microtubule-associated protein 2 and NeuN (+) immunoreactivities, and attenuated calpain activation in both the cortex and the hippocampus at 24 h after the injury; the reduction in neurodegeneration remained evident up to 14 days post-injury. In synaptosomal uptake assays, MS-153 up-regulated GLT-1 activity in the naïve rat brain but did not reverse the reduced activity of GLT-1 in traumatically-injured brains. This study demonstrates that administration of MS-153 in the acute post-traumatic period provides acute and long-term neuroprotection for TBI and suggests that the neuroprotective effects of MS-153 are related to mechanisms other than GLT-1 activation, such as the inhibition of voltage-gated calcium channels.
Collapse
Affiliation(s)
| | - Douglas P. Fox
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Argie Zoubroulis
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Ole Valente Mortensen
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Ramesh Raghupathi
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Abstract
OBJECTIVES The paradoxical development of chronic abdominal pain is an underrecognized side effect of opioid use. Narcotic bowel syndrome (NBS), occurring in a small proportion of chronic opioid users, consists of chronic or intermittent abdominal pain, which often increases in severity despite continued or escalating dosages of opioids prescribed to relieve pain. METHODS A PubMed search was conducted using terms such as "narcotic bowel syndrome" and "opioid hyperalgesia" through January 2014. RESULTS Abdominal pain is the defining symptom of NBS and is thought to be mediated by central nervous system dysfunction; it should be distinguished from the peripheral side effects of opioids, such as nausea, bloating, intermittent vomiting, abdominal distension, and constipation. This latter cluster of symptoms is called opioid bowel dysfunction, although it may co-occur with NBS. Hypothesized mechanisms of the central effects of opioids on nociception in NBS include spinal cord inflammation and dysfunction in opioid receptor activity and related neuroanatomical substrates. With continued use, ∼6% of patients taking narcotics chronically will develop NBS, with profound consequences in terms of daily function. The primary management paradigm for NBS is a structured opioid withdrawal program accompanied by centrally acting adjunctive therapy comprising antidepressants, benzodiazepines, and clonidine to target pain, anxiety, and depression, and prevent withdrawal effects, in addition to peripherally acting agents such as laxatives (e.g., osmotic laxatives and chloride channel activators) to control transient constipation. Such structured withdrawal programs have been prospectively evaluated in small clinical trials and have met with considerable success in the short term. CONCLUSIONS Because rates of NBS are likely to rise, integrated intensive pharmacotherapy and psychosocial interventions are needed to help patients with NBS go off and stay off opioids. These programs will likely also reduce comorbid psychopathology and lead to adequate pain control and improved quality of life.
Collapse
|
30
|
Sari Y, Toalston JE, Rao PSS, Bell RL. Effects of ceftriaxone on ethanol, nicotine or sucrose intake by alcohol-preferring (P) rats and its association with GLT-1 expression. Neuroscience 2016; 326:117-125. [PMID: 27060486 DOI: 10.1016/j.neuroscience.2016.04.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 03/30/2016] [Accepted: 04/01/2016] [Indexed: 11/17/2022]
Abstract
Increased glutamatergic neurotransmission appears to mediate the reinforcing properties of drugs of abuse, including ethanol (EtOH). We have shown that administration of ceftriaxone (CEF), a β-lactam antibiotic, reduced EtOH intake and increased glutamate transporter 1 (GLT-1) expression in mesocorticolimbic regions of male and female alcohol-preferring (P) rats. In the present study, we tested whether CEF administration would reduce nicotine (NIC) and/or EtOH intake by adult female P rats. P rats were randomly assigned to 4 groups: (a) 5% sucrose (SUC) or 10% SUC [SUC], (b) 5% SUC+0.07mg/ml NIC and 10% SUC+0.14mg/ml NIC [NIC-SUC], 15% EtOH and 30% EtOH [EtOH] and (d) 15% EtOH+0.07mg/ml NIC and 30% EtOH+0.14mg/ml NIC [NIC-EtOH]. After achieving stable intakes (4weeks), the rats were administered 7 consecutive, daily i.p. injections of either saline or 200mg/kg CEF. The effects of CEF on intake were significant but differed across the reinforcers; such that ml/kg/day SUC was reduced by ∼30%, mg/kg/day NIC was reduced by ∼70% in the NIC-SUC group and ∼40% in the EtOH-NIC group, whereas g/kg/day EtOH was reduced by ∼40% in both the EtOH and EtOH-NIC group. The effects of CEF on GLT-1 expression were also studied. We found that CEF significantly increased GLT-1 expression in the prefrontal cortex and the nucleus accumbens of the NIC and NIC-EtOH rats as compared to NIC and NIC-EtOH saline-treated rats. These findings provide further support for GLT-1-associated mechanisms in EtOH and/or NIC abuse. The present results along with previous reports of CEF's efficacy in reducing cocaine self-administration in rats suggest that modulation of GLT-1 expression and/or activity is an important pharmacological target for treating polysubstance abuse and dependence.
Collapse
Affiliation(s)
- Youssef Sari
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology, Toledo, OH 43614, USA.
| | - Jamie E Toalston
- Department of Psychiatry and Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - P S S Rao
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology, Toledo, OH 43614, USA
| | - Richard L Bell
- Department of Psychiatry and Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
31
|
Roberts-Wolfe DJ, Kalivas PW. Glutamate Transporter GLT-1 as a Therapeutic Target for Substance Use Disorders. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2016; 14:745-56. [PMID: 26022265 DOI: 10.2174/1871527314666150529144655] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 05/18/2015] [Indexed: 12/13/2022]
Abstract
The development of new treatments for substance use disorders requires identification of targetable molecular mechanisms. Pathology in glutamatergic neurotransmission system in brain reward circuitry has been implicated in relapse to multiple classes of drugs. Glutamate transporter 1 (GLT-1) crucially regulates glutamatergic signaling by removing excess glutamate from the extrasynaptic space. The purpose of this review is to highlight the effects of addictive drug use on GLT-1 and glutamate uptake, and using GLT-1 as a target in addiction pharmacotherapy. Cocaine, opioids, ethanol, nicotine, amphetamines, and cannabinoids each affect GLT-1 expression and glutamate uptake, and restoring GLT-1 expression with N-acetylcysteine or ceftriaxone shows promise in correcting pre-clinical and clinical manifestations of drug addiction.
Collapse
Affiliation(s)
- Douglas J Roberts-Wolfe
- Department of Neuroscience, Medical University of So Carolina, 173 Ashley Ave, BSB403, Charleston, SC 29425, USA.
| | | |
Collapse
|
32
|
D'Souza MS. Glutamatergic transmission in drug reward: implications for drug addiction. Front Neurosci 2015; 9:404. [PMID: 26594139 PMCID: PMC4633516 DOI: 10.3389/fnins.2015.00404] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/12/2015] [Indexed: 12/12/2022] Open
Abstract
Individuals addicted to drugs of abuse such as alcohol, nicotine, cocaine, and heroin are a significant burden on healthcare systems all over the world. The positive reinforcing (rewarding) effects of the above mentioned drugs play a major role in the initiation and maintenance of the drug-taking habit. Thus, understanding the neurochemical mechanisms underlying the reinforcing effects of drugs of abuse is critical to reducing the burden of drug addiction in society. Over the last two decades, there has been an increasing focus on the role of the excitatory neurotransmitter glutamate in drug addiction. In this review, pharmacological and genetic evidence supporting the role of glutamate in mediating the rewarding effects of the above described drugs of abuse will be discussed. Further, the review will discuss the role of glutamate transmission in two complex heterogeneous brain regions, namely the nucleus accumbens (NAcc) and the ventral tegmental area (VTA), which mediate the rewarding effects of drugs of abuse. In addition, several medications approved by the Food and Drug Administration that act by blocking glutamate transmission will be discussed in the context of drug reward. Finally, this review will discuss future studies needed to address currently unanswered gaps in knowledge, which will further elucidate the role of glutamate in the rewarding effects of drugs of abuse.
Collapse
Affiliation(s)
- Manoranjan S D'Souza
- Pharmaceutical and Biomedical Sciences, Raabe College of Pharmacy, Ohio Northern University Ada, OH, USA
| |
Collapse
|
33
|
Fontana ACK. Current approaches to enhance glutamate transporter function and expression. J Neurochem 2015; 134:982-1007. [DOI: 10.1111/jnc.13200] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 05/19/2015] [Accepted: 05/20/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Andréia C. K. Fontana
- Department of Pharmacology and Physiology; Drexel University College of Medicine; Philadelphia Pennsylvania USA
| |
Collapse
|
34
|
Koyama Y. Functional alterations of astrocytes in mental disorders: pharmacological significance as a drug target. Front Cell Neurosci 2015. [PMID: 26217185 PMCID: PMC4491615 DOI: 10.3389/fncel.2015.00261] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Astrocytes play an essential role in supporting brain functions in physiological and pathological states. Modulation of their pathophysiological responses have beneficial actions on nerve tissue injured by brain insults and neurodegenerative diseases, therefore astrocytes are recognized as promising targets for neuroprotective drugs. Recent investigations have identified several astrocytic mechanisms for modulating synaptic transmission and neural plasticity. These include altered expression of transporters for neurotransmitters, release of gliotransmitters and neurotrophic factors, and intercellular communication through gap junctions. Investigation of patients with mental disorders shows morphological and functional alterations in astrocytes. According to these observations, manipulation of astrocytic function by gene mutation and pharmacological tools reproduce mental disorder-like behavior in experimental animals. Some drugs clinically used for mental disorders affect astrocyte function. As experimental evidence shows their role in the pathogenesis of mental disorders, astrocytes have gained much attention as drug targets for mental disorders. In this paper, I review functional alterations of astrocytes in several mental disorders including schizophrenia, mood disorder, drug dependence, and neurodevelopmental disorders. The pharmacological significance of astrocytes in mental disorders is also discussed.
Collapse
Affiliation(s)
- Yutaka Koyama
- Laboratory of Pharmacology, Faculty of Pharmacy, Osaka Ohtani University Tondabayashi, Osaka, Japan
| |
Collapse
|
35
|
Ceftriaxone attenuates acquisition and facilitates extinction of cocaine-induced suppression of saccharin intake in C57BL/6J mice. Physiol Behav 2015; 149:174-80. [PMID: 26066719 DOI: 10.1016/j.physbeh.2015.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 05/08/2015] [Accepted: 06/06/2015] [Indexed: 12/14/2022]
Abstract
Growing evidence implicates glutamate homeostasis in a number of behaviors observed in addiction such as acquisition of drug taking, motivation, and reinstatement. To date, however, the role of glutamate homeostasis in the avoidance of natural rewards due to exposure to drugs of abuse has received little attention. The aim of the current study was to evaluate the beta-lactam antibiotic, ceftriaxone, which has been shown to normalize disrupted glutamate homeostasis associated with exposure to drugs of abuse, in cocaine-induced suppression of saccharin intake in C57BL/6J mice. Briefly, C57BL/6J mice received daily injections of either 200mg/kg ceftriaxone or saline. Mice were then given access to 0.15% saccharin for 1h and immediately injected intraperitoneally with either saline or 30 mg/kg cocaine; taste-drug pairings occurred every 24h for 5 trials followed by a final CS only trial. One week following taste-drug pairings, extinction was evaluated in a series of one- and two-bottle saccharin intake tests. Individual differences in cocaine-induced suppression were observed (i.e., low and high suppressors) with differential effects of ceftriaxone. Ceftriaxone delayed suppression of saccharin intake in high suppressors but prevented suppression in low suppressors. In addition, ceftriaxone history facilitated extinction in the high suppressors. These data suggest that changes in glutamate homeostasis may be involved in the formation and expression of cocaine-induced suppression of saccharin intake in mice.
Collapse
|
36
|
Trivedi MS, Deth R. Redox-based epigenetic status in drug addiction: a potential contributor to gene priming and a mechanistic rationale for metabolic intervention. Front Neurosci 2015; 8:444. [PMID: 25657617 PMCID: PMC4302946 DOI: 10.3389/fnins.2014.00444] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 12/16/2014] [Indexed: 12/26/2022] Open
Abstract
Alcohol and other drugs of abuse, including psychostimulants and opioids, can induce epigenetic changes: a contributing factor for drug addiction, tolerance, and associated withdrawal symptoms. DNA methylation is a major epigenetic mechanism and it is one of more than 200 methylation reactions supported by methyl donor S-adenosylmethionine (SAM). Levels of SAM are controlled by cellular redox status via the folate and vitamin B12-dependent enzyme methionine synthase (MS). For example, under oxidative conditions MS is inhibited, diverting its substrate homocysteine (HCY) to the trans sulfuration pathway. Alcohol, dopamine, and morphine, can alter intracellular levels of glutathione (GSH)-based cellular redox status, subsequently affecting SAM levels and DNA methylation status. Here, existing evidence is presented in a coherent manner to propose a novel hypothesis implicating the involvement of redox-based epigenetic changes in drug addiction. Further, we discuss how a “gene priming” phenomenon can contribute to the maintenance of redox and methylation status homeostasis under various stimuli including drugs of abuse. Additionally, a new mechanistic rationale for the use of metabolic interventions/redox-replenishers as symptomatic treatment of alcohol and other drug addiction and associated withdrawal symptoms is also provided. Hence, the current review article strengthens the hypothesis that neuronal metabolism has a critical bidirectional coupling with epigenetic changes in drug addiction exemplified by the link between redox-based metabolic changes and resultant epigenetic consequences under the effect of drugs of abuse.
Collapse
Affiliation(s)
- Malav S Trivedi
- Department of Pharmaceutical Sciences, Northeastern University Boston, MA, USA
| | - Richard Deth
- Department of Pharmaceutical Sciences, Northeastern University Boston, MA, USA
| |
Collapse
|
37
|
Aal-Aaboda M, Alhaddad H, Osowik F, Nauli SM, Sari Y. Effects of (R)-(-)-5-methyl-1-nicotinoyl-2-pyrazoline on glutamate transporter 1 and cysteine/glutamate exchanger as well as ethanol drinking behavior in male, alcohol-preferring rats. J Neurosci Res 2015; 93:930-7. [PMID: 25601490 DOI: 10.1002/jnr.23554] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 12/09/2014] [Accepted: 12/16/2014] [Indexed: 12/20/2022]
Abstract
Alcohol consumption is largely associated with alterations in the extracellular glutamate concentrations in several brain reward regions. We recently showed that glutamate transporter 1 (GLT-1) is downregulated following chronic exposure to ethanol for 5 weeks in alcohol-preferring (P) rats and that upregulation of the GLT-1 levels in nucleus accumbens and prefrontal cortex results, in part, in attenuating ethanol consumption. Cystine glutamate antiporter (xCT) is also downregulated after chronic ethanol exposure in P rats, and its upregulation could be valuable in attenuating ethanol drinking. This study examines the effect of a synthetic compound, (R)-(-)-5-methyl-1-nicotinoyl-2-pyrazoline (MS-153), on ethanol drinking and expressions of GLT-1 and xCT in the amygdala and the hippocampus of P rats. P rats were exposed to continuous free-choice access to water, 15% and 30% ethanol, and food for 5 weeks, after which they received treatments of MS-153 or vehicle for 5 days. The results show that MS-153 treatment significantly reduces ethanol consumption. It was revealed that GLT-1 and xCT expressions were downregulated in both the amygdala and the hippocampus of ethanol-vehicle-treated rats (ethanol-vehicle group) compared with water-control animals. MS-153 treatment upregulated GLT-1 and xCT expressions in these brain regions. These findings demonstrate an important role for MS-153 in these glutamate transporters for the attenuation of ethanol-drinking behavior.
Collapse
Affiliation(s)
- Munaf Aal-Aaboda
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, Ohio
| | | | | | | | | |
Collapse
|
38
|
Alhaddad H, Kim NT, Aal-Aaboda M, Althobaiti YS, Leighton J, Boddu SHS, Wei Y, Sari Y. Effects of MS-153 on chronic ethanol consumption and GLT1 modulation of glutamate levels in male alcohol-preferring rats. Front Behav Neurosci 2014; 8:366. [PMID: 25400560 PMCID: PMC4214358 DOI: 10.3389/fnbeh.2014.00366] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 10/05/2014] [Indexed: 12/26/2022] Open
Abstract
We have recently shown that upregulation of glutamate transporter 1 (GLT1) in the brain is associated in part with reduction in ethanol intake in alcohol-preferring (P) male rats. In this study, we investigated the effects of a synthetic compound, (R)-(−)-5-methyl-1-nicotinoyl-2-pyrazoline (MS-153), known to activate GLT1 on ethanol consumption as well as GLT1 expression and certain signaling pathways in P rats. P rats were given 24-h concurrent access to 15 and 30% ethanol, water and food for 5 weeks. On week 6, P rats received MS-153 at a dose of 50 mg/kg (i.p.) or a vehicle (i.p.) for 5 consecutive days. We also tested the effect of MS-153 on daily sucrose (10%) intake. Our studies revealed a significant decrease in ethanol intake at the dose of 50 mg/kg MS-153 from Day 1 through 14. In addition, MS-153 at dose of 50 mg/kg did not induce any significant effect on sucrose intake. Importantly, we found that MS-153 upregulated the GLT1 level in the nucleus accumbens (NAc) but not in the prefrontal cortex (PFC). In accordance, we found upregulation of nuclear NFkB-65 level in NAc in MS-153-treated group, however, IkBα was downregulated in MS-153-treated group in NAc. We did not find any changes in NFkB-65 and IkBα levels in PFC. Interestingly, we revealed that p-Akt was downregulated in ethanol vehicle treated groups in the NAc; this downregulation was reversed by MS-153 treatment. We did not observe any significant differences in glutamate aspartate transporter (GLAST) expression among all groups. These findings reveal MS-153 as a GLT1 modulator that may have potential as a therapeutic drug for the treatment of alcohol dependence.
Collapse
Affiliation(s)
- Hasan Alhaddad
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Toledo, OH, USA
| | - Nathaniel T Kim
- Department of Chemistry, Columbia University New York, NY, USA
| | - Munaf Aal-Aaboda
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Toledo, OH, USA
| | - Yusuf S Althobaiti
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Toledo, OH, USA
| | - James Leighton
- Department of Chemistry, Columbia University New York, NY, USA
| | - Sai H S Boddu
- Department of Pharmacy Practice, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Toledo, OH, USA
| | - Yangjie Wei
- Department of Pharmacy Practice, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Toledo, OH, USA
| | - Youssef Sari
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Toledo, OH, USA
| |
Collapse
|
39
|
Hillemacher T, Leggio L, Heberlein A. Investigational therapies for the pharmacological treatment of alcoholism. Expert Opin Investig Drugs 2014; 24:17-30. [PMID: 25164385 DOI: 10.1517/13543784.2014.954037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Alcohol dependence is one of the most important psychiatric disorders leading to enormous harm in individuals and indeed within society. Yet, although alcohol dependence is a disease of significant importance, the availability of efficacious pharmacological treatment is still limited. Areas covered: The current review focuses on neurobiological pathways that are the rationale for recent preclinical and clinical studies testing novel compounds that could be used as treatments for alcohol dependence. These neurobiological mechanisms include the: glutamatergic, dopaminergic and GABA mediated pathways as well as neuroendocrine systems. There is also an interest in the approaches for influencing chromatin structure. Expert opinion: There are several compounds in Phase I and Phase II clinical studies that have produced potentially useful results for the treating alcoholism. Further evaluation is still necessary, and the implementation of Phase III studies will help to elucidate the usefulness of these compounds. It is important that personalized approaches (e.g., pharmacogenomics) are investigated in these later studies, as the efficacy of different compounds may vary substantially between subgroups of patients.
Collapse
Affiliation(s)
- Thomas Hillemacher
- Hannover Medical School, Center for Addiction Research (CARe), Department of Psychiatry, Social Psychiatry and Psychotherapy , Carl-Neuberg-Str. 1, 30625 Hannover , Germany +49 511 532 2427 ; +49 511 532 2415 ;
| | | | | |
Collapse
|
40
|
Inhibition of vesicular glutamate transporters contributes to attenuate methamphetamine-induced conditioned place preference in rats. Behav Brain Res 2014; 267:1-5. [DOI: 10.1016/j.bbr.2014.02.047] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/19/2014] [Accepted: 02/22/2014] [Indexed: 11/23/2022]
|
41
|
Sarkar A, Rakshit S, Chall S, Mati SS, Singharoy D, Bañuelos J, López Arbeloa I, Bhattacharya SC. Micellar charge induced emissive response of a bio-active 3-pyrazolyl-2-pyrazoline derivative: a spectroscopic and quantum chemical analysis. RSC Adv 2014. [DOI: 10.1039/c4ra06497f] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
HOMO–LUMO distribution of PYZ in its ground and first singly excited state.
Collapse
Affiliation(s)
- Arindam Sarkar
- Department of Chemistry
- Jadavpur University
- Kolkata-700032, India
| | | | - Sayantani Chall
- Department of Chemistry
- Jadavpur University
- Kolkata-700032, India
| | | | - Dipti Singharoy
- Department of Chemistry
- Jadavpur University
- Kolkata-700032, India
| | - Jorge Bañuelos
- Departamento de Química Física
- Facultad de Cienciasy Tecnología
- Universidad del País Vasco-EHU
- 48080-Bilbao, Spain
| | - Iñigo López Arbeloa
- Departamento de Química Física
- Facultad de Cienciasy Tecnología
- Universidad del País Vasco-EHU
- 48080-Bilbao, Spain
| | | |
Collapse
|
42
|
Beardsley PM, Hauser KF. Glial modulators as potential treatments of psychostimulant abuse. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 69:1-69. [PMID: 24484974 DOI: 10.1016/b978-0-12-420118-7.00001-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Glia (including astrocytes, microglia, and oligodendrocytes), which constitute the majority of cells in the brain, have many of the same receptors as neurons, secrete neurotransmitters and neurotrophic and neuroinflammatory factors, control clearance of neurotransmitters from synaptic clefts, and are intimately involved in synaptic plasticity. Despite their prevalence and spectrum of functions, appreciation of their potential general importance has been elusive since their identification in the mid-1800s, and only relatively recently have they been gaining their due respect. This development of appreciation has been nurtured by the growing awareness that drugs of abuse, including the psychostimulants, affect glial activity, and glial activity, in turn, has been found to modulate the effects of the psychostimulants. This developing awareness has begun to illuminate novel pharmacotherapeutic targets for treating psychostimulant abuse, for which targeting more conventional neuronal targets has not yet resulted in a single, approved medication. In this chapter, we discuss the molecular pharmacology, physiology, and functional relationships that the glia have especially in the light in which they present themselves as targets for pharmacotherapeutics intended to treat psychostimulant abuse disorders. We then review a cross section of preclinical studies that have manipulated glial processes whose behavioral effects have been supportive of considering the glia as drug targets for psychostimulant-abuse medications. We then close with comments regarding the current clinical evaluation of relevant compounds for treating psychostimulant abuse, as well as the likelihood of future prospects.
Collapse
Affiliation(s)
| | - Kurt F Hauser
- Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
43
|
Niciu MJ, Henter ID, Sanacora G, Zarate CA. Glial abnormalities in substance use disorders and depression: does shared glutamatergic dysfunction contribute to comorbidity? World J Biol Psychiatry 2014; 15:2-16. [PMID: 24024876 PMCID: PMC4180366 DOI: 10.3109/15622975.2013.829585] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Preclinical and clinical research in neuropsychiatric disorders, particularly mood and substance use disorders, have historically focused on neurons; however, glial cells-astrocytes, microglia, and oligodendrocytes - also play key roles in these disorders. METHODS Peer-reviewed PubMed/Medline articles published through December 2012 were identified using the following keyword combinations: glia, astrocytes, oligodendrocytes/glia, microglia, substance use, substance abuse, substance dependence, alcohol, opiate, opioid, cocaine, psychostimulants, stimulants, and glutamate. RESULTS Depressive and substance use disorders are highly comorbid, suggesting a common or overlapping aetiology and pathophysiology. Reduced astrocyte cell number occurs in both disorders. Altered glutamate neurotransmission and metabolism - specifically changes in the levels/activity of transporters, receptors, and synaptic proteins potentially related to synaptic physiology - appear to be salient features of both disorders. Glial cell pathology may also underlie the pathophysiology of both disorders via impaired astrocytic production of neurotrophic factors. Microglial/neuroinflammatory pathology is also evident in both depressive and substance use disorders. Finally, oligodendrocyte impairment decreases myelination and impairs expression of myelin-related genes in both substance use and depressive disorders. CONCLUSIONS Glial-mediated glutamatergic dysfunction is a common neuropathological pathway in both substance use and depression. Therefore, glutamatergic neuromodulation is a rational drug target in this comorbidity.
Collapse
Affiliation(s)
- Mark J. Niciu
- Yale University Department of Psychiatry/Connecticut Mental Health Center (CMHC), Clinical Neuroscience Research Unit (CNRU), New Haven, CT, USA,Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, Bethesda, MD, USA
| | - Ioline D. Henter
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD USA
| | - Gerard Sanacora
- Yale University Department of Psychiatry/Connecticut Mental Health Center (CMHC), Clinical Neuroscience Research Unit (CNRU), New Haven, CT, USA
| | - Carlos A. Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, Bethesda, MD, USA
| |
Collapse
|
44
|
Wu Q, Xia S, Lin J, Cao D, Chen W, Liu L, Fu Y, Liang J, Cao M. Effects of the altered activity of δ-opioid receptor on the expression of glutamate transporter type 3 induced by chronic exposure to morphine. J Neurol Sci 2013; 335:174-81. [PMID: 24120272 DOI: 10.1016/j.jns.2013.09.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 09/17/2013] [Accepted: 09/18/2013] [Indexed: 11/29/2022]
Abstract
Altered δ-opioid receptor (DOR) activity can affect the activity and function of excitatory amino acid transporter 3 (EAAT3), but the effects of DOR on EAAT3 expression in morphine relapse remain unknown. In this study, a C6δ cell line and SD rats in a conditioned place preference (CPP) reinstatement model were used. Here, we show that EAAT3 protein levels in C6δ cells decreased significantly after chronic exposure to morphine (10 μM) for 48 h and returned to normal 12 h after drug withdrawal. When C6δ cells were re-exposed to 5 μM morphine for 4 h, EAAT3 protein levels again decreased significantly. The selective μ opioid receptor (MOR) specific agonist DAMGO had a similar effect as morphine, and CTOP, a specific MOR blocker, reversed the declined expression of EAAT3 protein triggered by morphine exposure. The selective DOR agonist [d-pen2, 5] enkephalin (DPDPE) significantly increased EAAT3 expression in C6δ cells and even reversed the decreased EAAT3 expression caused by chronic morphine exposure. The non specific antagonist naloxone, but not the DOR inhibitor Naltrindole (NTI), reversed the decreased EAAT3 expression in C6δ cells caused by chronic morphine exposure. In vivo, EAAT3 levels in the prefrontal cortex of rats with morphine-induced CPP reinstatement significantly decreased. Naloxone completely suppressed reinstatement and reversed the decrease in EAAT3 expression induced by morphine re-exposure. In contrast, NTI only weakened CPP reinstatement and exerted no influence on EAAT3 expression. These findings suggest that DOR can affect the expression of EAAT3. However, the morphine-induced down-regulation of EAAT3 in C6δ cells and in the prefrontal cortex of rats may not be mediated by DOR.
Collapse
Affiliation(s)
- Qiang Wu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hutchinson MR, Watkins LR. Why is neuroimmunopharmacology crucial for the future of addiction research? Neuropharmacology 2013; 76 Pt B:218-27. [PMID: 23764149 DOI: 10.1016/j.neuropharm.2013.05.039] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 05/13/2013] [Accepted: 05/23/2013] [Indexed: 12/13/2022]
Abstract
A major development in drug addiction research in recent years has been the discovery that immune signaling within the central nervous system contributes significantly to mesolimbic dopamine reward signaling induced by drugs of abuse, and hence is involved in the presentation of reward behaviors. Additionally, in the case of opioids, these hypotheses have advanced through to the discovery of the novel site of opioid action at the innate immune pattern recognition receptor Toll-like receptor 4 as the necessary triggering event that engages this reward facilitating central immune signaling. Thus, the hypothesis of major proinflammatory contributions to drug abuse was born. This review will examine these key discoveries, but also address several key lingering questions of how central immune signaling is able to contribute in this fashion to the pharmacodynamics of drugs of abuse. It is hoped that by combining the collective wisdom of neuroscience, immunology and pharmacology, into Neuroimmunopharmacology, we may more fully understanding the neuronal and immune complexities of how drugs of abuse, such as opioids, create their rewarding and addiction states. Such discoveries will point us in the direction such that one day soon we might successfully intervene to successfully treat drug addiction. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
Affiliation(s)
- Mark R Hutchinson
- Discipline of Physiology, School of Medical Sciences, University of Adelaide, Level 5, Medical School South, Frome Rd, Adelaide, South Australia 5005, Australia.
| | | |
Collapse
|
46
|
Rao PSS, Sari Y. Glutamate transporter 1: target for the treatment of alcohol dependence. Curr Med Chem 2013; 19:5148-56. [PMID: 22680643 DOI: 10.2174/092986712803530511] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 05/04/2012] [Accepted: 05/07/2012] [Indexed: 12/19/2022]
Abstract
Emerging evidence indicates that many aspects of alcohol and drug dependence involve changes in glutamate transmission. A number of studies have reported that drugs of abuse, including alcohol and cocaine, alter glutamate transport. Extracellular glutamate is regulated by a number of glutamate transporters in various brain regions. Of these transporters, glutamate transporter (GLT1) is a key player in the removal of most of the extracellular glutamate. Similar to neurodegenerative disease models, in which there is dysfunction of the glutamatergic excitatory system, the role of GLT1 has been tested in drug dependence models that show dysfunction of glutamate transmission. We and others have recently found that ceftriaxone, an FDA-approved drug known to elevate GLT1 expression, attenuates cue-induced cocaine relapse. Moreover, we recently found that alcohol-preferring rats treated with ceftriaxone showed a significant dosedependent reduction in alcohol consumption. We also demonstrated that ceftriaxone-induced upregulation of GLT1 expression was associated with increases in glutamate uptake in Huntington's disease mouse model. Importantly, ceftriaxone is currently in clinical trials for the treatment of amyotrophic lateral sclerosis. This review provides information about the potential therapeutic role of GLT1 for the treatment of alcohol abuse and dependence.
Collapse
Affiliation(s)
- P S S Rao
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology, Health Science Campus, 3000 Arlington Avenue, Toledo, OH 43614, USA
| | | |
Collapse
|
47
|
Sari Y. Potential therapeutic role of glutamate transporter 1 for the treatment of alcohol dependence. ACTA ACUST UNITED AC 2013; 1:6. [PMID: 24409344 DOI: 10.13172/2053-0285-1-1-574] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Evidence has demonstrated that deficits in glutamate transmission impair neurocircuits involved in drug abuse or drug-seeking behaviour and affect many aspects of neuroplasticity associated with alcohol and drug addiction. Alcohol-seeking behaviour is promoted by increased glutamate transmission in key regions of the mesocorticolimbic reward circuit, including the nucleus accumbens and prefrontal cortex. Glutamate transmission or glutamate uptake is regulated by a number of glutamate transporters in the brain regions. Among these glutamate transporters, glutamate transporter 1 (GLT1; its human homolog is the excitatory amino acid transporter 2, EAAT2) regulates the removal of majority of the extracellular glutamate. The role of GLT1 has been tested in alcohol and other drugs of abuse models with dysfunction in glutamate transmission. We recently reported that treatment of alcohol-preferring rats with compounds ceftriaxone and GPI-1046, known to upregulate GLT1 levels, showed reduction in alcohol intake and attenuation of relapse-like ethanol-drinking behaviour. Furthermore, we demonstrated that upregulation of GLT1 was associated with attenuation of cue-induced cocaine relapse. Together, we suggest that GLT1 is considered as a potential therapeutic target for the treatment of drug dependence, including alcohol. The aim of this critical review was to discuss the potential therapeutic role of GLT1 for the treatment of alcohol dependence. CONCLUSION Dysfunction of glutamate transmission has been suggested to impair neurocircuits involved in alcohol dependence, which affect neuroplasticity that is associated with ethanol intake.
Collapse
Affiliation(s)
- Y Sari
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology, Toledo, Ohio, USA
| |
Collapse
|
48
|
Sari Y, Franklin KM, Alazizi A, Rao PSS, Bell RL. Effects of ceftriaxone on the acquisition and maintenance of ethanol drinking in peri-adolescent and adult female alcohol-preferring (P) rats. Neuroscience 2013; 241:229-38. [PMID: 23537837 DOI: 10.1016/j.neuroscience.2013.03.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 02/15/2013] [Accepted: 03/07/2013] [Indexed: 10/27/2022]
Abstract
Increased glutamatergic neurotransmission appears to mediate the reinforcing properties of drugs of abuse, including ethanol (EtOH). We recently reported that the administration of ceftriaxone (CEF), a β-lactam antibiotic known to upregulate glutamate transporter 1 (GLT1) levels/activity, decreased the maintenance of EtOH intake in adult male alcohol-preferring (P) rats. In the present study, we tested whether CEF administration would reduce the acquisition and maintenance of EtOH drinking in adolescent and adult female P rats. The rats were treated with saline or 200mg/kg ceftriaxone for 7 days (starting at 35 or 75 days old, respectively) followed by the EtOH acquisition test. Five weeks later the effects of CEF were examined regarding the maintenance of EtOH intake. For the maintenance test, half of the animals that received CEF during acquisition received CEF for 7 days and the other half received saline for 7 days. Saline-treated acquisition animals were treated similarly. The results indicated that pretreatment with ceftriaxone reduced the maintenance of EtOH intake in both animals that started as adolescents and those that started as adults. However, the beneficial effect of CEF was more pronounced in rats pretreated with CEF as adults compared with rats pretreated as adolescents. Reductions in EtOH intake by ceftriaxone were paralleled by an upregulation of GLT1 protein levels in both the nucleus accumbens (∼25% in rats starting at both ages) and prefrontal cortex (∼50% in rats starting as peri-adolescents and ∼65% in those starting as adults). These findings provide further support for GLT1-associated mechanisms in high alcohol-consuming behavior, and hold promise for the development of effective treatments targeting alcohol abuse and dependence.
Collapse
Affiliation(s)
- Y Sari
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology, Toledo, OH 43614, USA.
| | | | | | | | | |
Collapse
|
49
|
Sari Y, Sreemantula SN. Neuroimmunophilin GPI-1046 reduces ethanol consumption in part through activation of GLT1 in alcohol-preferring rats. Neuroscience 2012; 227:327-35. [PMID: 23059796 PMCID: PMC3505992 DOI: 10.1016/j.neuroscience.2012.10.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 10/01/2012] [Accepted: 10/02/2012] [Indexed: 01/13/2023]
Abstract
We have previously shown that ceftriaxone, β-lactam antibiotic known to upregulate glutamate transporter 1 (GLT1), reduced ethanol intake in alcohol-preferring (P) rats. GLT1 is a glial glutamate transporter that regulates the majority of extracellular glutamate uptake. We tested in this study the effects of neuroimmunophilin GPI-1046 (3-(3-pyridyl)-1-propyl (2S)-1-(3,3-dimethyl-1,2-dioxopentyl)-2-pyrrolidinecarboxylate), known also to upregulate GLT1 expression, in ethanol intake in P rats. Male P rats had concurrent access to free choice of 15% and 30% ethanol, water, and food for five weeks. On Week 6, P rats continued in this drinking and food regimen and they were administered either 10 or 20mg/kg GPI-1046 (i.p.), or a vehicle for five consecutive days. Body weight, ethanol intake, and water consumption were measured daily for 8 days starting on Day 1 of GPI-1046 or vehicle i.p. injections. We have also tested the effect of GPI-1046 (20mg/kg) on daily sucrose (10%) intake. The data revealed significant dose-dependent effects in the reduction of ethanol intake starting 48 h after the first treatment with GPI-1046 throughout treatment and post-treatment periods. There were also dose-dependent increases in water intake. However, GPI-1046 treatment did not affect the body weight of all animals nor sucrose intake. Importantly, GPI-1046 (20mg/kg) increased GLT1 level compared to all groups in nucleus accumbens core (NAc-core). Alternatively, GPI-1046 (10mg/kg) upregulated GLT1 level in NAc-core compared to vehicle (ethanol naïve) group. Moreover, both doses of GPI-1046 increased significantly GLT1 level in the prefrontal cortex (PFC) compared to ethanol naïve vehicle group. GPI-1046 (20mg/kg) increased GLT1 level in PFC compared to naïve control group that was exposed to water and food only. These findings demonstrated that neuroimmunophilin GPI-1046 attenuates ethanol intake in part through the upregulation of GLT1 in PFC and NAc-core.
Collapse
Affiliation(s)
- Y Sari
- University of Toledo, College of Pharmacy and Pharmaceutical Sciences, Department of Pharmacology, Toledo, OH 43614, United States.
| | | |
Collapse
|
50
|
Kufahl PR, Hood LE, Nemirovsky NE, Barabas P, Halstengard C, Villa A, Moore E, Watterson LR, Olive MF. Positive Allosteric Modulation of mGluR5 Accelerates Extinction Learning but Not Relearning Following Methamphetamine Self-Administration. Front Pharmacol 2012. [PMID: 23189054 PMCID: PMC3506114 DOI: 10.3389/fphar.2012.00194] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Recent studies have implicated glutamate neurotransmission as an important substrate for the extinction of conditioned behaviors, including responding for drug reinforcement. Positive allosteric modulation of the type-5 metabotropic glutamate receptor (mGluR5) in particular has emerged as a treatment strategy for the enhancement of extinction of drug-motivated behaviors. Here, we investigated the effects of the mGluR5 positive allosteric modulator CDPPB, a compound known for its cognitive enhancing effects in rodents, on extinction learning in rats with different histories of methamphetamine (METH) training. Rats were trained to self-administer METH under two conditions: 16 daily sessions of short access (90 min/day, ShA), or eight daily sessions of short access followed by eight sessions of long access (6 h/day, LgA). Control rats self-administered sucrose pellets in daily 30 min sessions. Next, rats were administered vehicle or 30 mg/kg CDPPB prior to seven consecutive daily extinction sessions, subjected to additional extinction sessions to re-establish a post-treatment baseline, and then tested for reinstatement of behavior in the presence of METH- or sucrose-paired cues. Rats were then subjected to a second series of extinction sessions, preceded by vehicle or 30 mg/kg CDPPB, and an additional test for cue-triggered reinstatement. CDPPB treatment resulted in a more rapid extinction of responding on the active lever, especially in the early sessions of the first extinction sequence. However, treatment effects were minimal during subsequent cue reinstatement tests and non-existent during the second series of extinction sessions. Rats with histories of ShA, LgA, and sucrose training expressed similar behavioral sensitivities to CDPPB, with LgA rats demonstrating a modestly higher treatment effect. Positive allosteric modulation of mGluR5 may therefore have some beneficial effects on efforts to facilitate extinction learning and reduce methamphetamine seeking.
Collapse
Affiliation(s)
- Peter R Kufahl
- Department of Psychology, Arizona State University Tempe, AZ, USA
| | | | | | | | | | | | | | | | | |
Collapse
|