1
|
Li P, Lei W, Dong Y, Wang X, Ye X, Tian Y, Yang Y, Liu J, Li N, Niu X, Wang X, Tian Y, Xu L, Yang Y, Liu J. mGluR7: The new player protecting the central nervous system. Ageing Res Rev 2024; 102:102554. [PMID: 39454762 DOI: 10.1016/j.arr.2024.102554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/16/2024] [Accepted: 10/20/2024] [Indexed: 10/28/2024]
Abstract
Metabotropic glutamate receptor 7 (mGluR7) belongs to the family of type III mGluR receptor, playing an important part in the central nervous system (CNS) through response to neurotransmitter regulation, reduction of excitatory toxicity, and early neuronal development. Drugs targeting mGluR7 (mGluR7 agonists, antagonists, and allosteric modulators) may be among the most promising agents for the treatment of CNS disorders, such as psychiatric disorders, neurodegenerative diseases, and neurodevelopmental impairments, though these potential therapies are at early stages and the data are still limited. In this review, we summarized the structure and function of mGluR7 and discussed recent progress on mGluR7 agonists and antagonists. A deeper understanding of mGluR7 will contribute to uncovering the molecular mechanisms of neuroprotection and providing a theoretical basis for the formulation of therapeutic strategies.
Collapse
Affiliation(s)
- Pan Li
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China; Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Ophthalmology, Xi'an No.1 Hospital, Faculty of Life Sciences and Medicine, Northwest University, 30 Fenxiang Road, Xi'an 710002, China
| | - Wangrui Lei
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yushu Dong
- Department of Neurosurgery, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Xiaowu Wang
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Xingyan Ye
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Ye Tian
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yaru Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Jie Liu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Ning Li
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Xiaochen Niu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Xin Wang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yifan Tian
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Lu Xu
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yang Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an 710069, China.
| | - Jincheng Liu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
2
|
Kahvandi N, Ebrahimi Z, Sharifi M, Karimi SA, Shahidi S, Salehi I, Haddadi R, Sarihi A. S-3,4-DCPG, a potent orthosteric agonist for the mGlu8 receptor, facilitates extinction and inhibits the reinstatement of morphine-induced conditioned place preference in male rats. Pharmacol Biochem Behav 2024; 240:173772. [PMID: 38653345 DOI: 10.1016/j.pbb.2024.173772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
The limbic system, particularly the NAc, shows a high concentration of metabotropic glutamate receptors (mGluRs). Recent evidence suggests the significant involvement of mGluRs in mental disorders, including substance abuse and addiction. The objective of this study was to examine the involvement of mGlu8 receptors in the NAc in the mechanisms underlying the extinction and reinstatement of conditioned place preference (CPP) induced by morphine. Male Wistar rats underwent surgical implantation of bilateral cannulas in the NAc and were assessed in a CPP protocol. In study 1 at the same time as the extinction phase, the rats were given varying doses of S-3,4-DCPG (0.03, 0.3, and 3 μg/0.5 μl). In study 2, rats that had undergone CPP extinction were given S-3,4-DCPG (0.03, 0.3, and 3 μg/0.5 μl) five minutes prior to receiving a subthreshold dose of morphine (1 mg/kg) in order to reactivate the previously extinguished morphine response. The findings demonstrated that administering S-3,4-DCPG directly into the accumbens nucleus resulted in a decrease in the duration of the CPP extinction phase. Moreover, dose-dependent administration of S-3,4-DCPG into the NAc inhibited CPP reinstatement. The observations imply that microinjection of S-3,4-DCPG as a potent orthosteric agonist with high selectivity for the mGlu8 receptor into the NAc promotes the process of extinction while concurrently exerting inhibitory effects on the reinstatement of morphine-induced CPP. This effect may be associated with the modulation of glutamate engagement within the NAc and the plasticity of reward pathways at the synaptic level.
Collapse
Affiliation(s)
- Nazanin Kahvandi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Physiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Zahra Ebrahimi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Physiology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Sharifi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Asaad Karimi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Salehi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasool Haddadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abdolrahman Sarihi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
3
|
Soares-Cunha C, Heinsbroek JA. Ventral pallidal regulation of motivated behaviors and reinforcement. Front Neural Circuits 2023; 17:1086053. [PMID: 36817646 PMCID: PMC9932340 DOI: 10.3389/fncir.2023.1086053] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023] Open
Abstract
The interconnected nuclei of the ventral basal ganglia have long been identified as key regulators of motivated behavior, and dysfunction of this circuit is strongly implicated in mood and substance use disorders. The ventral pallidum (VP) is a central node of the ventral basal ganglia, and recent studies have revealed complex VP cellular heterogeneity and cell- and circuit-specific regulation of reward, aversion, motivation, and drug-seeking behaviors. Although the VP is canonically considered a relay and output structure for this circuit, emerging data indicate that the VP is a central hub in an extensive network for reward processing and the regulation of motivation that extends beyond classically defined basal ganglia borders. VP neurons respond temporally faster and show more advanced reward coding and prediction error processing than neurons in the upstream nucleus accumbens, and regulate the activity of the ventral mesencephalon dopamine system. This review will summarize recent findings in the literature and provide an update on the complex cellular heterogeneity and cell- and circuit-specific regulation of motivated behaviors and reinforcement by the VP with a specific focus on mood and substance use disorders. In addition, we will discuss mechanisms by which stress and drug exposure alter the functioning of the VP and produce susceptibility to neuropsychiatric disorders. Lastly, we will outline unanswered questions and identify future directions for studies necessary to further clarify the central role of VP neurons in the regulation of motivated behaviors. Significance: Research in the last decade has revealed a complex cell- and circuit-specific role for the VP in reward processing and the regulation of motivated behaviors. Novel insights obtained using cell- and circuit-specific interrogation strategies have led to a major shift in our understanding of this region. Here, we provide a comprehensive review of the VP in which we integrate novel findings with the existing literature and highlight the emerging role of the VP as a linchpin of the neural systems that regulate motivation, reward, and aversion. In addition, we discuss the dysfunction of the VP in animal models of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Carina Soares-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Jasper A. Heinsbroek
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
4
|
Jensen KL, Jensen SB, Madsen KL. A mechanistic overview of approaches for the treatment of psychostimulant dependence. Front Pharmacol 2022; 13:854176. [PMID: 36160447 PMCID: PMC9493975 DOI: 10.3389/fphar.2022.854176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Psychostimulant use disorder is a major health issue around the world with enormous individual, family-related and societal consequences, yet there are no effective pharmacological treatments available. In this review, a target-based overview of pharmacological treatments toward psychostimulant addiction will be presented. We will go through therapeutic approaches targeting different aspects of psychostimulant addiction with focus on three major areas; 1) drugs targeting signalling, and metabolism of the dopamine system, 2) drugs targeting either AMPA receptors or metabotropic glutamate receptors of the glutamate system and 3) drugs targeting the severe side-effects of quitting long-term psychostimulant use. For each of these major modes of intervention, findings from pre-clinical studies in rodents to clinical trials in humans will be listed, and future perspectives of the different treatment strategies as well as their potential side-effects will be discussed. Pharmaceuticals modulating the dopamine system, such as antipsychotics, DAT-inhibitors, and disulfiram, have shown some promising results. Cognitive enhancers have been found to increase aspects of behavioural control, and drugs targeting the glutamate system such as modulators of metabotropic glutamate receptors and AMPA receptors have provided interesting changes in relapse behaviour. Furthermore, CRF-antagonists directed toward alleviating the symptoms of the withdrawal stage have been examined with interesting resulting changes in behaviour. There are promising results investigating therapeutics for psychostimulant addiction, but further preclinical work and additional human studies with a more stratified patient selection are needed to prove sufficient evidence of efficacy and tolerability.
Collapse
|
5
|
Luessen DJ, Conn PJ. Allosteric Modulators of Metabotropic Glutamate Receptors as Novel Therapeutics for Neuropsychiatric Disease. Pharmacol Rev 2022; 74:630-661. [PMID: 35710132 PMCID: PMC9553119 DOI: 10.1124/pharmrev.121.000540] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors, a family of G-protein-coupled receptors, have been identified as novel therapeutic targets based on extensive research supporting their diverse contributions to cell signaling and physiology throughout the nervous system and important roles in regulating complex behaviors, such as cognition, reward, and movement. Thus, targeting mGlu receptors may be a promising strategy for the treatment of several brain disorders. Ongoing advances in the discovery of subtype-selective allosteric modulators for mGlu receptors has provided an unprecedented opportunity for highly specific modulation of signaling by individual mGlu receptor subtypes in the brain by targeting sites distinct from orthosteric or endogenous ligand binding sites on mGlu receptors. These pharmacological agents provide the unparalleled opportunity to selectively regulate neuronal excitability, synaptic transmission, and subsequent behavioral output pertinent to many brain disorders. Here, we review preclinical and clinical evidence supporting the utility of mGlu receptor allosteric modulators as novel therapeutic approaches to treat neuropsychiatric diseases, such as schizophrenia, substance use disorders, and stress-related disorders.
Collapse
|
6
|
Kupchik YM, Prasad AA. Ventral pallidum cellular and pathway specificity in drug seeking. Neurosci Biobehav Rev 2021; 131:373-386. [PMID: 34562544 DOI: 10.1016/j.neubiorev.2021.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/03/2021] [Accepted: 09/05/2021] [Indexed: 01/12/2023]
Abstract
The ventral pallidum (VP) is central to the reinforcing effects across a variety of drugs and relapse to drug seeking. Emerging studies from animal models of reinstatement reveal a complex neurobiology of the VP that contributes to different aspects of relapse to drug seeking. This review builds on classical understanding of the VP as part of the final common pathway of relapse but also discusses the properties of the VP as an independent structure. These include VP neural anatomical subregions, cellular heterogeneity, circuitry, neurotransmitters and peptides. Collectively, this review provides a current understanding of the VP from molecular to circuit level architecture that contributes to both the appetitive and aversive symptoms of drug addiction. We show the complex neurobiology of the VP in drug seeking, emphasizing its critical role in addiction, and review strategic approaches that target the VP to reduce relapse rates.
Collapse
Affiliation(s)
- Yonatan M Kupchik
- Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem. P.O. Box 12271, Jerusalem, 9112102, Israel
| | - Asheeta A Prasad
- School of Psychology, UNSW Sydney, NSW, 2052, Australia; Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia.
| |
Collapse
|
7
|
Nall RW, Heinsbroek JA, Nentwig TB, Kalivas PW, Bobadilla AC. Circuit selectivity in drug versus natural reward seeking behaviors. J Neurochem 2021; 157:1450-1472. [PMID: 33420731 PMCID: PMC8178159 DOI: 10.1111/jnc.15297] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/16/2020] [Accepted: 01/03/2021] [Indexed: 12/23/2022]
Abstract
Substance use disorder (SUD) is characterized, in part by behavior biased toward drug use and away from natural sources of reward (e.g., social interaction, food, sex). The neurobiological underpinnings of SUDs reveal distinct brain regions where neuronal activity is necessary for the manifestation of SUD-characteristic behaviors. Studies that specifically examine how these regions are involved in behaviors motivated by drug versus natural reward allow determinations of which regions are necessary for regulating seeking of both reward types, and appraisals of novel SUD therapies for off-target effects on behaviors motivated by natural reward. Here, we evaluate studies directly comparing regulatory roles for specific brain regions in drug versus natural reward. While it is clear that many regions drive behaviors motivated by all reward types, based on the literature reviewed we propose a set of interconnected regions that become necessary for behaviors motivated by drug, but not natural rewards. The circuitry is selectively necessary for drug seeking includes an Action/Reward subcircuit, comprising nucleus accumbens, ventral pallidum, and ventral tegmental area, a Prefrontal subcircuit comprising prelimbic, infralimbic, and insular cortices, a Stress subcircuit comprising the central nucleus of the amygdala and the bed nucleus of the stria terminalis, and a Diencephalon circuit including lateral hypothalamus. Evidence was mixed for nucleus accumbens shell, insular cortex, and ventral pallidum. Studies for all other brain nuclei reviewed supported a necessary role in regulating both drug and natural reward seeking. Finally, we discuss emerging strategies to further disambiguate the necessity of brain regions in drug- versus natural reward-associated behaviors.
Collapse
Affiliation(s)
- Rusty W. Nall
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Jasper A. Heinsbroek
- Department of Anesthesiology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Todd B. Nentwig
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Peter W. Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
- These authors share senior authorship
| | - Ana-Clara Bobadilla
- School of Pharmacy, University of Wyoming, Laramie, WY, USA
- These authors share senior authorship
| |
Collapse
|
8
|
De Sa Nogueira D, Bourdy R, Filliol D, Quessada C, McCort-Tranchepain I, Acher F, Zwiller J, Romieu P, Befort K. LSP2-9166, an orthosteric mGlu4 and mGlu7 receptor agonist, reduces cocaine self-administration under a progressive ratio schedule in rats. Neurosci Lett 2020; 764:135603. [PMID: 33387661 DOI: 10.1016/j.neulet.2020.135603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 11/24/2022]
Abstract
Cocaine addiction is a serious health issue in Western countries. Despite the regular increase in cocaine consumption across the population, there is no specific treatment for cocaine addiction. Critical roles for glutamate neurotransmission in the rewarding effects of psychostimulants as well as relapse have been suggested and accumulating evidence indicates that targeting mGlu group III receptors could represent a promising strategy to develop therapeutic compounds to treat addiction. In this context, the aim of our study was to examine the effect of LSP2-9166, a mGlu4/mGlu7 receptor orthosteric agonist, on the motivation for cocaine intake. We used an intravenous self-administration paradigm in male Wistar rats as a reliable model of voluntary drug intake. We first evaluated the direct impact of cocaine on Grm4 and Grm7 gene expression. Voluntary cocaine intake under a fixed ratio schedule of injections induced an increase of both mGlu4 and mGlu7 receptor transcripts in nucleus accumbens and hippocampus. We then evaluated the ability of LSP2-9166 to affect cocaine self-administration under a progressive ratio schedule of reinforcement. We found that this compound inhibits the motivation to obtain the drug, although it induced a hypolocomotor effect which could biais motivation index. Our findings demonstrate that mGlu group III receptors represent new targets for decreasing motivation to self-administer cocaine.
Collapse
Affiliation(s)
- David De Sa Nogueira
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France
| | - Romain Bourdy
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France
| | - Dominique Filliol
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France
| | - Cyril Quessada
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France; Université de Strasbourg, Inserm, UMR-S1118, 11 rue Humann, F-67000, Strasbourg, France
| | - Isabelle McCort-Tranchepain
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR8601, Université de Paris 45 rue des Saints-Pères, F-75270, Paris Cedex 6, France
| | - Francine Acher
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR8601, Université de Paris 45 rue des Saints-Pères, F-75270, Paris Cedex 6, France
| | - Jean Zwiller
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France
| | - Pascal Romieu
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France
| | - Katia Befort
- Université de Strasbourg, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Faculté de Psychologie, UMR 7364, CNRS 12 rue Goethe, F-67000, Strasbourg, France.
| |
Collapse
|
9
|
Gregory KJ, Goudet C. International Union of Basic and Clinical Pharmacology. CXI. Pharmacology, Signaling, and Physiology of Metabotropic Glutamate Receptors. Pharmacol Rev 2020; 73:521-569. [PMID: 33361406 DOI: 10.1124/pr.119.019133] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors respond to glutamate, the major excitatory neurotransmitter in the mammalian brain, mediating a modulatory role that is critical for higher-order brain functions such as learning and memory. Since the first mGlu receptor was cloned in 1992, eight subtypes have been identified along with many isoforms and splice variants. The mGlu receptors are transmembrane-spanning proteins belonging to the class C G protein-coupled receptor family and represent attractive targets for a multitude of central nervous system disorders. Concerted drug discovery efforts over the past three decades have yielded a wealth of pharmacological tools including subtype-selective agents that competitively block or mimic the actions of glutamate or act allosterically via distinct sites to enhance or inhibit receptor activity. Herein, we review the physiologic and pathophysiological roles for individual mGlu receptor subtypes including the pleiotropic nature of intracellular signal transduction arising from each. We provide a comprehensive analysis of the in vitro and in vivo pharmacological properties of prototypical and commercially available orthosteric agonists and antagonists as well as allosteric modulators, including ligands that have entered clinical trials. Finally, we highlight emerging areas of research that hold promise to facilitate rational design of highly selective mGlu receptor-targeting therapeutics in the future. SIGNIFICANCE STATEMENT: The metabotropic glutamate receptors are attractive therapeutic targets for a range of psychiatric and neurological disorders. Over the past three decades, intense discovery efforts have yielded diverse pharmacological tools acting either competitively or allosterically, which have enabled dissection of fundamental biological process modulated by metabotropic glutamate receptors and established proof of concept for many therapeutic indications. We review metabotropic glutamate receptor molecular pharmacology and highlight emerging areas that are offering new avenues to selectively modulate neurotransmission.
Collapse
Affiliation(s)
- Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| | - Cyril Goudet
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| |
Collapse
|
10
|
Masugi-Tokita M, Tomita K, Kobayashi K, Yoshida T, Kageyama S, Sakamoto H, Kawauchi A. Metabotropic Glutamate Receptor Subtype 7 Is Essential for Ejaculation. Mol Neurobiol 2020; 57:5208-5218. [PMID: 32865662 DOI: 10.1007/s12035-020-02090-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/24/2020] [Indexed: 01/27/2023]
Abstract
Metabotropic glutamate receptor subtype 7 (mGluR7) is a member of the group III mGluRs, which are negatively coupled to adenylate cyclase via Gi/Go proteins and localized to presynaptic active zones of the mammalian central nervous system (CNS). To elucidate the mechanism of impaired reproductivity of mGluR7 knockout (KO) mice, we investigated sexual behavior in this line, which exhibits ejaculatory disorder, although with normal sexual motivation and erectile function. To identify the site of action within the CNS responsible for the effect of mGluR7 on ejaculation, we then used a para-chloroamphetamine (PCA)-induced ejaculation model. Intrathecal administration of the mGluR7-selective antagonist 6-(4-methoxyphenyl)-5-methyl-3-pyridin-4-ylisoxazolo[4,5-c]pyridin-4(5H)-one (MMPIP) into the lumbosacral spinal cord inhibited PCA-induced ejaculation. Immunohistochemistry revealed mGluR7-like immunoreactivity (LI) expressed in the same area where lumbar spinothalamic (LSt) cells regulate the parasympathetic ejaculatory pathway. At high magnification, the apposition of mGluR7-LI puncta and neuronal nitric oxide synthase (nNOS)-LI-positive putative parasympathetic preganglionic neurons was evident. These results indicate that mGluR7 in the lumbosacral spinal cord regulates ejaculation by potentiating the excitability of parasympathetic preganglionic neurons. The ejaculatory disorder is a major issue in the field of male reproductive function. Erectile dysfunction (ED) can be treated by phosphodiesterase type 5 inhibitors like sildenafil (Viagra®), but the ejaculatory disorder cannot. Lack of understanding of the ejaculatory mechanism hinders the development of therapies for ejaculatory problems. This study is the first to demonstrate that mGluR7 regulates ejaculation and the results provide insight into the mechanism of ejaculation as well as a strategy for future therapies to treat ejaculatory disorders in humans.
Collapse
Affiliation(s)
- Miwako Masugi-Tokita
- Department of Urology, Shiga University of Medical Science, Otsu, Japan. .,World Premier International Research Center Initiative-Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, Japan. .,Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.
| | - Keiji Tomita
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| | - Kenichi Kobayashi
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| | - Tetsuya Yoshida
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| | - Susumu Kageyama
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| | - Hirotaka Sakamoto
- Ushimado Marine Institute (UMI), Graduate School of Natural Science and Technology, Okayama University, Setouchi, Japan
| | - Akihiro Kawauchi
- Department of Urology, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
11
|
Xu L, Nan J, Lan Y. The Nucleus Accumbens: A Common Target in the Comorbidity of Depression and Addiction. Front Neural Circuits 2020; 14:37. [PMID: 32694984 PMCID: PMC7338554 DOI: 10.3389/fncir.2020.00037] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/22/2020] [Indexed: 12/21/2022] Open
Abstract
The comorbidity of depression and addiction has become a serious public health issue, and the relationship between these two disorders and their potential mechanisms has attracted extensive attention. Numerous studies have suggested that depression and addiction share common mechanisms and anatomical pathways. The nucleus accumbens (NAc) has long been considered a key brain region for regulating many behaviors, especially those related to depression and addiction. In this review article, we focus on the association between addiction and depression, highlighting the potential mediating role of the NAc in this comorbidity via the regulation of changes in the neural circuits and molecular signaling. To clarify the mechanisms underlying this association, we summarize evidence from overlapping reward neurocircuitry, the resemblance of cellular and molecular mechanisms, and common treatments. Understanding the interplay between these disorders should help guide clinical comorbidity prevention and the search for a new target for comorbidity treatment.
Collapse
Affiliation(s)
- Le Xu
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University School of Medicine, Yanji City, China
| | - Jun Nan
- Department of Orthopedics, Affiliated Hospital of Yanbian University, Yanji City, China
| | - Yan Lan
- Department of Physiology and Pathophysiology, College of Medicine, Yanbian University School of Medicine, Yanji City, China
| |
Collapse
|
12
|
Boccella S, Marabese I, Guida F, Luongo L, Maione S, Palazzo E. The Modulation of Pain by Metabotropic Glutamate Receptors 7 and 8 in the Dorsal Striatum. Curr Neuropharmacol 2020; 18:34-50. [PMID: 31210112 PMCID: PMC7327935 DOI: 10.2174/1570159x17666190618121859] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/01/2019] [Accepted: 05/31/2019] [Indexed: 12/28/2022] Open
Abstract
The dorsal striatum, apart from controlling voluntary movement, displays a recently demonstrated pain inhibition. It is connected to the descending pain modulatory system and in particular to the rostral ventromedial medulla through the medullary dorsal reticular nucleus. Diseases of the basal ganglia, such as Parkinson's disease, in addition to being characterized by motor disorders, are associated with pain and hyperactivation of the excitatory transmission. A way to counteract glutamatergic hyperactivation is through the activation of group III metabotropic glutamate receptors (mGluRs), which are located on presynaptic terminals inhibiting neurotransmitter release. So far the mGluRs of group III have been the least investigated, owing to a lack of selective tools. More recently, selective ligands for each mGluR of group III, in particular positive and negative allosteric modulators, have been developed and the role of each subtype is starting to emerge. The neuroprotective potential of group III mGluRs in pathological conditions, such as those characterized by elevate glutamate, has been recently shown. In the dorsal striatum, mGluR7 and mGluR8 are located at glutamatergic corticostriatal terminals and their stimulation inhibits pain in pathological conditions such as neuropathic pain. The two receptors in the dorsal striatum have instead a different role in pain control in normal conditions. This review will discuss recent results focusing on the contribution of mGluR7 and mGluR8 in the dorsal striatal control of pain. The role of mGluR4, whose antiparkinsonian activity is widely reported, will also be addressed.
Collapse
Affiliation(s)
- Serena Boccella
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Enza Palazzo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| |
Collapse
|
13
|
Cid JM, Lavreysen H, Tresadern G, Pérez-Benito L, Tovar F, Fontana A, Trabanco AA. Computationally Guided Identification of Allosteric Agonists of the Metabotropic Glutamate 7 Receptor. ACS Chem Neurosci 2019; 10:1043-1054. [PMID: 30216043 DOI: 10.1021/acschemneuro.8b00331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The metabotropic glutamate 7 (mGlu7) receptor belongs to the group III of mGlu receptors. Since the mGlu7 receptor can control excitatory neurotransmission in the hippocampus and cortex, modulation of the receptor may have therapeutic benefit in several CNS diseases. However, mGlu7 remains relatively unexplored among the eight known mGlu receptors partly because of the limited availability of tool compounds to interrogate its potential therapeutic utility. Here we report the discovery of a new class of mGlu7 allosteric agonists. Hits originating from virtual screening were followed up with further analogue searching and screening, leading to a novel series of mGlu7 allosteric agonists. Guided by docking into a structural model of the mGlu7 receptor the initial hit 5 was successfully optimized to analogues with comparable potencies and more attractive drug-like attributes than AMN082.
Collapse
Affiliation(s)
- Jose María Cid
- Janssen Research and Development, Calle Jarama 75A, Toledo 45007, Spain
| | - Hilde Lavreysen
- Janssen Research and Development, Turnhoutseweg 30, 2440 Beerse, Belgium
| | - Gary Tresadern
- Janssen Research and Development, Turnhoutseweg 30, 2440 Beerse, Belgium
| | - Laura Pérez-Benito
- Laboratori de Medicina Computacional Unitat de Bioestadistica, Facultat de Medicina, Universitat Autonoma de Barcelona, Bellaterra 08193, Spain
| | - Fulgencio Tovar
- Villapharma Research
S.L., Parque Tecnológico de Fuente Álamo. Ctra. El Estrecho-Lobosillo, Km. 2.5- Av. Azul, 30320 Fuente Álamo de Murcia, Murcia, Spain
| | - Alberto Fontana
- Janssen Research and Development, Calle Jarama 75A, Toledo 45007, Spain
| | | |
Collapse
|
14
|
Caprioli D, Justinova Z, Venniro M, Shaham Y. Effect of Novel Allosteric Modulators of Metabotropic Glutamate Receptors on Drug Self-administration and Relapse: A Review of Preclinical Studies and Their Clinical Implications. Biol Psychiatry 2018; 84:180-192. [PMID: 29102027 PMCID: PMC5837933 DOI: 10.1016/j.biopsych.2017.08.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 12/31/2022]
Abstract
Results from preclinical rodent studies during the last 20 years implicated glutamate neurotransmission in different brain regions in drug self-administration and rodent models of relapse. These results, along with evidence for drug-induced neuroadaptations in glutamatergic neurons and receptors, suggested that addiction might be treatable by medications that inhibit glutamatergic responses to drugs of abuse, drug-associated cues, and stressors. This idea is supported by findings in rodent and primate models that drug self-administration and relapse are reduced by systemic injections of antagonists of ionotropic glutamate receptors or metabotropic glutamate receptors (mGluRs) or orthosteric agonists of mGluR2/3. However, these compounds have not advanced to clinical use because of potential side effects and other factors. This state of affairs has led to the development of positive allosteric modulators (PAMs) and negative allosteric modulators (NAMs) of mGluRs. PAMs and NAMs of mGluRs, either of which can inhibit evoked glutamate release, may be suitable for testing in humans. We reviewed results from recent studies of systemically injected PAMs and NAMs of mGluRs in rodents and monkeys, focusing on whether they reduce drug self-administration, reinstatement of drug seeking, and incubation of drug craving. We also review results from rat studies in which PAMs or NAMs of mGluRs were injected intracranially to reduce drug self-administration and reinstatement. We conclude that PAMs and NAMs of mGluRs should be considered for clinical trials.
Collapse
Affiliation(s)
- Daniele Caprioli
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy.
| | - Zuzana Justinova
- Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD, USA
| | - Marco Venniro
- Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD, USA
| | - Yavin Shaham
- Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD, USA
| |
Collapse
|
15
|
Vatankhah M, Sarihi A, Komaki A, Shahidi S, Haghparast A. AMN082—a metabotropic glutamate receptor type 7 allosteric agonist in the NAc facilitates extinction and inhibits the reinstatement of morphine-induced conditioned place preference in male rats. Brain Res Bull 2018; 140:28-33. [DOI: 10.1016/j.brainresbull.2018.03.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 11/27/2022]
|
16
|
Vatankhah M, Karimi-Haghighi S, Sarihi A, Haghparast A. Intra-accumbal administration of AMN082, a metabotropic glutamate receptor type 7 allosteric agonist, inhibits the acquisition but not the expression of morphine-induced conditioned place preference in rats. Neurosci Lett 2018; 681:56-61. [PMID: 29800675 DOI: 10.1016/j.neulet.2018.05.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/20/2018] [Accepted: 05/21/2018] [Indexed: 11/17/2022]
Abstract
The nucleus accumbens (NAc) plays a primary role in opioid reward. The actions of glutamate are mediated by the activation of ionotropic and metabotropic glutamate receptors (mGluRs). Previous documents have shown the extensive distributions of the different types of mGluRs, including mGluR7, in regions that are involved in opioid reward, such as the NAc. In this study, seventy male Wistar rats were used to investigate the role of mGluR7 receptors in the NAc on the acquisition and expression of morphine-induced conditioned place preference (CPP). In Experiment 1, to determine the effect of AMN082, a selective mGluR7 allosteric agonist, on the acquisition of morphine-induced conditioned place preference (CPP), the rats bilaterally received AMN082 (1, 3 and 5 μg/0.5 μL DMSO) during three-day conditioning by morphine (5 mg/kg). In Experiment 2, the rats bilaterally received AMN082 (5 μg/0.5 μL DMSO) 5 min prior to the post-conditioning test to investigate the effect of AMN082 on the expression of morphine-induced CPP. The results showed that the intra-accumbal injection of AMN082 prevents the acquisition of morphine-induced CPP in a dose-dependent manner. However, intra-accumbal injection of AMN082 had no effect on the expression of morphine-induced CPP. The findings propose that the mGluR7 in the NAc inhibits the acquisition of morphine-induced CPP that could be mediated by inhibition of NMDA receptors in the NAc.
Collapse
Affiliation(s)
- Mahsaneh Vatankhah
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saeideh Karimi-Haghighi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, PO Box 19615-1178, Tehran, Iran
| | - Abdolrahman Sarihi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, PO Box 19615-1178, Tehran, Iran.
| |
Collapse
|
17
|
Hajasova Z, Canestrelli C, Acher F, Noble F, Marie N. Role of mGlu7 receptor in morphine rewarding effects is uncovered by a novel orthosteric agonist. Neuropharmacology 2018; 131:424-430. [PMID: 29307544 DOI: 10.1016/j.neuropharm.2018.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 12/26/2017] [Accepted: 01/02/2018] [Indexed: 11/26/2022]
Abstract
Opiate dependence is a major health issue and despite the existence of opioid substitution treatment, relapse frequently occurs. Group III metabotropic glutamate (mGlu) receptors has received much attention as a putative target in ethanol and cocaine addiction, but no data on opiate addiction exist. So we investigated the role of group III mGlu receptors in morphine rewarding effects through the expression and the reinstatement of conditioned place preference (CPP) using a newly synthesized mGlu4/mGlu7 receptor orthosteric agonist, LSP2-9166. We found that LSP2-9166 blocked morphine CPP expression and reinstatement after extinction. Blockade of CPP expression with LSP2-9166 was abolished when using XAP044, a mGlu7 antagonist. We also found that LSP2-9166 at the dose active for blocking morphine reward was devoid of any effect on locomotion, hedonic state, spatial memory, anxiety or depression. Altogether our data demonstrated that group III mGlu receptors, and more specifically mGlu7, might be a valuable target in opiate addiction.
Collapse
Affiliation(s)
- Zuzana Hajasova
- CNRS ERL 3649, Neuroplasticité et Thérapie des Addictions, Paris, France; INSERM UMR-S 1124, Paris, France; Université Paris Descartes, Paris, France
| | - Corinne Canestrelli
- CNRS ERL 3649, Neuroplasticité et Thérapie des Addictions, Paris, France; INSERM UMR-S 1124, Paris, France; Université Paris Descartes, Paris, France
| | - Francine Acher
- Université Paris Descartes, Paris, France; CNRS UMR8601, Laboratoire de Chimie & Biochimie Pharmacologiques et Toxicologiques, Paris, France
| | - Florence Noble
- CNRS ERL 3649, Neuroplasticité et Thérapie des Addictions, Paris, France; INSERM UMR-S 1124, Paris, France; Université Paris Descartes, Paris, France
| | - Nicolas Marie
- CNRS ERL 3649, Neuroplasticité et Thérapie des Addictions, Paris, France; INSERM UMR-S 1124, Paris, France; Université Paris Descartes, Paris, France.
| |
Collapse
|
18
|
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 2017; 68:816-71. [PMID: 27363441 DOI: 10.1124/pr.116.012484] [Citation(s) in RCA: 379] [Impact Index Per Article: 54.1] [Reference Citation Analysis] [Abstract] [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
|
19
|
Bahi A. Decreased anxiety, voluntary ethanol intake and ethanol-induced CPP acquisition following activation of the metabotropic glutamate receptor 8 “mGluR8”. Pharmacol Biochem Behav 2017; 155:32-42. [DOI: 10.1016/j.pbb.2017.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/14/2017] [Accepted: 03/16/2017] [Indexed: 10/19/2022]
|
20
|
Kahl E, Fendt M. Metabotropic Glutamate Receptors 7 within the Nucleus Accumbens are Involved in Relief Learning in Rats. Curr Neuropharmacol 2017; 14:405-12. [PMID: 27296637 PMCID: PMC4983753 DOI: 10.2174/1570159x13666150425002017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/03/2015] [Accepted: 04/22/2016] [Indexed: 11/26/2022] Open
Abstract
Relief learning is an appetitive association of a formally neutral cue with relief induced by the offset of an aversive stimulus. Since the nucleus accumbens mediates relief learning and accumbal metabotropic glutamate receptors 7 (mGluR7) modulate appetitive-like processes, we hypothesized that accumbal mGluR7 may be involved in the modulation of relief learning. Therefore, we injected the allosteric mGluR7 agonist AMN082 into the nucleus accumbens and tested the effects of these injections on acquisition and expression of relief memory, as well as on the reactivity to electric stimuli. AMN082 injections blocked acquisition but not expression of relief memory. In addition, accumbal AMN082 injections strongly reduced the locomotor reactivity to electric stimuli indicating antinociceptive effects. These antinociceptive effects might be causal for the blockade of relief learning after AMN082 injections. Taken together, the present study indicates that functional activation of accumbal mGluR7 has antinociceptive effects that interfere with relief learning.
Collapse
Affiliation(s)
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Leipziger Straße 44, D-39120 Magdeburg, Germany.
| |
Collapse
|
21
|
Melroy-Greif WE, Vadasz C, Kamens HM, McQueen MB, Corley RP, Stallings MC, Hopfer CJ, Krauter KS, Brown SA, Hewitt JK, Ehringer MA. Test for association of common variants in GRM7 with alcohol consumption. Alcohol 2016; 55:43-50. [PMID: 27788777 DOI: 10.1016/j.alcohol.2015.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 10/27/2015] [Accepted: 10/27/2015] [Indexed: 10/21/2022]
Abstract
Recent work using a mouse model has identified the glutamate metabotropic receptor 7 (Grm7) gene as a strong candidate gene for alcohol consumption. Although there has been some work examining the effect of human glutamate metabotropic receptor 7 (GRM7) polymorphisms on human substance use disorders, the majority of the work has focused on other psychiatric disorders such as ADHD, major depressive disorder, schizophrenia, bipolar disorder, panic disorder, and autism spectrum disorders. The current study aimed to evaluate evidence for association between GRM7 and alcohol behaviors in humans using a single nucleotide polymorphism (SNP) approach, as well as a gene-based approach. Using 1803 non-Hispanic European Americans (EAs) (source: the Colorado Center on Antisocial Drug Dependence [CADD]) and 1049 EA subjects from an independent replication sample (source: the Genetics of Antisocial Drug Dependence [GADD]), two SNPs in GRM7 were examined for possible association with alcohol consumption using two family-based association tests implemented in FBAT and QTDT. Rs3749380 was suggestively associated with alcohol consumption in the CADD sample (p = 0.010) with the minor T allele conferring risk. There was no evidence for association in the GADD sample. A gene-based test using four Genome-Wide Association Studies (GWAS) revealed no association between variation in GRM7 and alcohol consumption. This study had several limitations: the SNPs chosen likely do not tag expression quantitative trait loci; a human alcohol consumption phenotype was used, complicating the interpretation with respect to rodent studies that found evidence for a cis-regulatory link between alcohol preference and Grm7; and only common SNPs imputed in all four datasets were included in the gene-based test. These limitations highlight the fact that rare variants, some potentially important common signals in the gene, and regions farther upstream were not examined.
Collapse
|
22
|
Ahnaou A, Raeyemaekers L, Huysmans H, Drinkenburg W. Off-target potential of AMN082 on sleep EEG and related physiological variables: Evidence from mGluR7 (−/−) mice. Behav Brain Res 2016; 311:287-297. [DOI: 10.1016/j.bbr.2016.05.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 04/27/2016] [Accepted: 05/16/2016] [Indexed: 02/08/2023]
|
23
|
Li X, Markou A. Metabotropic Glutamate Receptor 7 (mGluR7) as a Target for the Treatment of Psychostimulant Dependence. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2016; 14:738-44. [PMID: 26022263 DOI: 10.2174/1871527314666150529145332] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 05/18/2015] [Indexed: 11/22/2022]
Abstract
Although few medications have been approved by the U.S. Food and Drug Administration (FDA) to assist people to quit tobacco smoking, there are no FDA-approved medications to treat dependence on other psychostimulant drugs, such as cocaine. The motivation to maintain psychostimulant drug seeking and self-administration involves alterations in glutamatergic neurotransmission. Thus, medications that modulate glutamate transmission may be effective treatments for psychostimulant dependence. One presynaptic inhibitory glutamate receptor that critically regulates glutamate transmission is the metabotropic glutamate 7 receptor (mGluR7). This review summarizes nonhuman experimental animal data that indicate a critical role for mGluR7 in drug-taking and drug-seeking behaviors for the psychostimulants cocaine and nicotine. AMN082, the only commercially available allosteric receptor agonist, has been used to investigate the role of mGluR7 in psychostimulant dependence. Systemic administration or microinjection of AMN082 into brain sites within the mesocorticolimbic system decreased self-administration and reinstatement of both cocaine and nicotine seeking. In vivo microdialysis results indicated that a nucleus accumbens-ventral pallidum γ-aminobutyric acid-ergic mechanism may underlie AMN082-induced antagonism of the reinforcing effects of cocaine, whereas a glutamate mGlu2/3 receptor mechanism underlies the AMN082-induced blockade of cocaine seeking. These findings indicate an important role for mGluR7 in mesolimbic areas in modulating the reinforcing effects of psychostimulant drugs, such as nicotine and cocaine, and the conditioned behaviors associated with drugs of abuse. Thus, selective mGluR7 agonists or positive allosteric modulators may have the potential to treat psychostimulant dependence.
Collapse
Affiliation(s)
| | - Athina Markou
- Department of Psychiatry, M/C 0603, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0603, USA.
| |
Collapse
|
24
|
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: 9.3] [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
|
25
|
Jang EY, Ryu YH, Lee BH, Chang SC, Yeo MJ, Kim SH, Folsom RJ, Schilaty ND, Kim KJ, Yang CH, Steffensen SC, Kim HY. Involvement of reactive oxygen species in cocaine-taking behaviors in rats. Addict Biol 2015; 20:663-75. [PMID: 24975938 DOI: 10.1111/adb.12159] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Reactive oxygen species (ROS) have been implicated in the development of behavioral sensitization following repeated cocaine exposure. We hypothesized that increased ROS following cocaine exposure would act as signaling molecules in the mesolimbic dopamine (DA) system, which might play an important role in mediating the reinforcing effects of cocaine. The aim of this study was to evaluate cocaine enhancement of brain metabolic activity and the effects of ROS scavengers on cocaine self-administration behavior, cocaine-induced ROS production in the nucleus accumbens (NAc) and cocaine enhancement of DA release in the NAc. Metabolic neural activity monitored by temperature and oxidative stress were increased in NAc following cocaine exposure. Systemic administration of the ROS scavenger N-tert-butyl-α-phenylnitrone (PBN) or 4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl (TEMPOL), either pre- or post-treatment, significantly decreased cocaine self-administration without affecting food intake. Infusion of TEMPOL into the NAc inhibited cocaine self-administration. Increased oxidative stress was found mainly on neurons, but not astrocytes, microglia or oligodendrocytes, in NAc of rats self-administering cocaine. TEMPOL significantly attenuated cocaine-induced enhancement of DA release in the NAc, compared to saline controls. TEMPOL had no effect on the enhancement of DA release produced by the DA transporter inhibitor GBR12909. Taken together, these findings suggest that enhancement of ROS production in NAc neurons contributes to the reinforcing effect of cocaine.
Collapse
Affiliation(s)
- Eun Young Jang
- College of Korean Medicine; Daegu Haany University; South Korea
- Department of Psychology; Center for Neuroscience; Brigham Young University; Provo UT USA
| | - Yeon-Hee Ryu
- Acupuncture, Moxibustion & Meridian Research Center; Division of Standard Research; Korea Institute of Oriental Medicine; South Korea
| | - Bong Hyo Lee
- College of Korean Medicine; Daegu Haany University; South Korea
| | - Su-Chan Chang
- College of Korean Medicine; Daegu Haany University; South Korea
| | - Mi Jin Yeo
- College of Korean Medicine; Daegu Haany University; South Korea
| | - Sang Hyun Kim
- College of Korean Medicine; Daegu Haany University; South Korea
| | - Ryan J. Folsom
- Department of Psychology; Center for Neuroscience; Brigham Young University; Provo UT USA
| | - Nathan D. Schilaty
- Department of Psychology; Center for Neuroscience; Brigham Young University; Provo UT USA
| | - Kwang Joong Kim
- College of Korean Medicine; Daegu Haany University; South Korea
| | - Chae Ha Yang
- College of Korean Medicine; Daegu Haany University; South Korea
| | - Scott C. Steffensen
- Department of Psychology; Center for Neuroscience; Brigham Young University; Provo UT USA
| | - Hee Young Kim
- College of Korean Medicine; Daegu Haany University; South Korea
| |
Collapse
|
26
|
Jenda M, Gawel K, Marszalek M, Komsta L, Kotlinska JH. AMN082, a metabotropic glutamate receptor 7 allosteric agonist, attenuates locomotor sensitization and cross-sensitization induced by cocaine and morphine in mice. Prog Neuropsychopharmacol Biol Psychiatry 2015; 57:166-75. [PMID: 25448778 DOI: 10.1016/j.pnpbp.2014.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 10/29/2014] [Accepted: 11/06/2014] [Indexed: 10/24/2022]
Abstract
Previous studies have indicated that metabotropic glutamate receptors 7 (mGluR7s) are involved in drug addiction. However, the role of these receptors in drug-induced behavioral sensitization is unknown. The aim of the present study was to determine whether systemic injection of AMN082, a selective mGluR7 allosteric agonist, reduces the cocaine- and morphine-induced hyperactivity and the development and expression of locomotor sensitization, and also affects the reciprocal cross-sensitization to the stimulant effect of cocaine and morphine in mice. AMN082 (1.25-10.0 mg/kg, i.p.) did not have an impact on locomotion of naive mice and did not affect the acute cocaine- or morphine-induced hyperactivity, except the dose of 10 mg/kg that suppressed the locomotor effect of both drugs. Repeated exposure to cocaine or morphine (10 mg/kg, 5× every 3 days) gradually increased locomotion during induction of sensitization and after 4 (cocaine) or 7 day (morphine) withdrawal phase when challenged with cocaine (10 mg/kg, i.p.) or morphine (10 mg/kg, i.p.) on day 17 or 20, respectively. Pretreatment of animals with the lower doses of AMN082 (1.25-5.0 mg/kg, i.p.), 30 min before every cocaine or morphine injection during repeated drug administration or before cocaine or morphine challenge, dose-dependently attenuated the development, as well as the expression of cocaine or morphine locomotor sensitization. AMN082 also inhibited the reciprocal cross-sensitization between these drugs. Prior to administration of MMPIP (10 mg/kg, i.p.), a selective mGluR7 antagonist reversed the inhibitory effect of AMN082 on the development or expression of cocaine or morphine sensitization. These data indicate that AMN082 attenuated the development and expression of cocaine and morphine sensitization, and the reciprocal cross-sensitization via a mechanism that involves mGluR7s. Thus, AMN082 might have therapeutic implications not only in the treatment of cocaine or opioid addiction but also in the treatment of cocaine/opioid polydrug-abusers.
Collapse
Affiliation(s)
- M Jenda
- Department of Pharmacology and Pharmacodynamics, Medical University, Lublin, Poland
| | - K Gawel
- Department of Pharmacology and Pharmacodynamics, Medical University, Lublin, Poland
| | - M Marszalek
- Department of Pharmacology and Pharmacodynamics, Medical University, Lublin, Poland
| | - L Komsta
- Department of Medicinal Chemistry, Medical University, Lublin, Poland
| | - J H Kotlinska
- Department of Pharmacology and Pharmacodynamics, Medical University, Lublin, Poland.
| |
Collapse
|
27
|
Kemppainen H, Nurmi H, Raivio N, Kiianmaa K. Enhanced Extracellular Glutamate and Dopamine in the Ventral Pallidum of Alcohol-Preferring AA and Alcohol-Avoiding ANA Rats after Morphine. Front Psychiatry 2015; 6:1. [PMID: 25653621 PMCID: PMC4299289 DOI: 10.3389/fpsyt.2015.00001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/01/2015] [Indexed: 11/20/2022] Open
Abstract
The purpose of the present study was to investigate the role of ventral pallidal opioidergic mechanisms in the control of ethanol intake by studying the effects of acute administration of morphine on the levels of GABA, glutamate, and dopamine in the ventral pallidum. The study was conducted using the alcohol-preferring Alko Alcohol (AA) and alcohol-avoiding Alko Non-Alcohol (ANA) rat lines that have well-documented differences in their voluntary ethanol intake and brain opioidergic systems. Therefore, examination of neurobiological differences between the lines is supposed to help to identify the neuronal mechanisms underlying ethanol intake, since selection pressure is assumed gradually to lead to enrichment of alleles promoting high or low ethanol intake, respectively. The effects of an acute dose of morphine (1 or 10 mg/kg s.c.) on the extracellular levels of GABA and glutamate in the ventral pallidum were monitored with in vivo microdialysis. The concentrations of GABA and glutamate in the dialyzates were determined with a high performance liquid chromatography system using fluorescent detection, while electrochemical detection was used for dopamine. The levels of glutamate in the rats injected with morphine 1 mg/kg were significantly above the levels found in the controls and in the rats receiving morphine 10 mg/kg. Morphine 10 mg/kg also increased the levels of dopamine. Morphine could not, however, modify the levels of GABA. The rat lines did not differ in any of the effects of morphine. The data suggest that the glutamatergic and dopaminergic systems in the ventral pallidum may mediate some effects of morphine. Since there were no differences between the AA and ANA lines, the basic hypothesis underlying the use of the genetic animal model suggests that the effects of morphine detected probably do not underlie the different intake of ethanol by the lines and contribute to the control of ethanol intake in these animals.
Collapse
Affiliation(s)
- Heidi Kemppainen
- Department of Alcohol, Drugs and Addiction, National Institute for Health and Welfare , Helsinki , Finland
| | - Harri Nurmi
- Department of Alcohol, Drugs and Addiction, National Institute for Health and Welfare , Helsinki , Finland
| | - Noora Raivio
- Department of Alcohol, Drugs and Addiction, National Institute for Health and Welfare , Helsinki , Finland
| | - Kalervo Kiianmaa
- Department of Alcohol, Drugs and Addiction, National Institute for Health and Welfare , Helsinki , Finland
| |
Collapse
|
28
|
Nucleus accumbens shell excitability is decreased by methamphetamine self-administration and increased by 5-HT2C receptor inverse agonism and agonism. Neuropharmacology 2014; 89:113-21. [PMID: 25229719 DOI: 10.1016/j.neuropharm.2014.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/25/2014] [Accepted: 09/01/2014] [Indexed: 11/21/2022]
Abstract
Methamphetamine profoundly increases brain monoamines and is a widely abused psychostimulant. The effects of methamphetamine self-administration on neuron function are not known for the nucleus accumbens, a brain region involved in addictive behaviors, including drug-seeking. One therapeutic target showing preclinical promise at attenuating psychostimulant-seeking is 5-HT2C receptors; however, the effects of 5-HT2C receptor ligands on neuronal physiology are unclear. 5-HT2C receptor agonism decreases psychostimulant-mediated behaviors, and the putative 5-HT2C receptor inverse agonist, SB 206553, attenuates methamphetamine-seeking in rats. To ascertain the effects of methamphetamine, and 5-HT2C receptor inverse agonism and agonism, on neuronal function in the nucleus accumbens, we evaluated methamphetamine, SB 206553, and the 5-HT2C receptor agonist and Ro 60-0175, on neuronal excitability within the accumbens shell subregion using whole-cell current-clamp recordings in forebrain slices ex vivo. We reveal that methamphetamine self-administration decreased generation of evoked action potentials. In contrast, SB 206553 and Ro 60-0175 increased evoked spiking, effects that were prevented by the 5-HT2C receptor antagonist, SB 242084. We also assessed signaling mechanisms engaged by 5-HT2C receptors, and determined that accumbal 5-HT2C receptors stimulated Gq, but not Gi/o. These findings demonstrate that methamphetamine-induced decreases in excitability of neurons within the nucleus accumbens shell were abrogated by both 5-HT2C inverse agonism and agonism, and this effect likely involved activation of Gq-mediated signaling pathways.
Collapse
|
29
|
Group III metabotropic glutamate receptors and drug addiction. Front Med 2014; 7:445-51. [PMID: 24078068 DOI: 10.1007/s11684-013-0291-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/05/2013] [Indexed: 02/06/2023]
Abstract
Neuroadaptations of glutamatergic transmission in the limbic reward circuitry are linked to persistent drug addiction. Accumulating data have demonstrated roles of ionotropic glutamate receptors and group I and II metabotropic glutamate receptors (mGluRs) in this event. Emerging evidence also identifies Gαi/o-coupled group III mGluRs (mGluR4/7/8 subtypes enriched in the limbic system) as direct substrates of drugs of abuse and active regulators of drug action. Auto- and heteroreceptors of mGluR4/7/8 reside predominantly on nerve terminals of glutamatergic corticostriatal and GABAergic striatopallidal pathways, respectively. These presynaptic receptors regulate basal and/or phasic release of respective transmitters to maintain basal ganglia homeostasis. In response to operant administration of common addictive drugs, such as psychostimulants (cocaine and amphetamine), alcohol and opiates, limbic group III mGluRs undergo drastic adaptations to contribute to the enduring remodeling of excitatory synapses and to usually suppress drug seeking behavior. As a result, a loss-of-function mutation (knockout) of individual group III receptor subtypes often promotes drug seeking. This review summarizes the data from recent studies on three group III receptor subtypes (mGluR4/7/8) expressed in the basal ganglia and analyzes their roles in the regulation of dopamine and glutamate signaling in the striatum and their participation in the addictive properties of three major classes of drugs (psychostimulants, alcohol, and opiates).
Collapse
|
30
|
Mercier MS, Lodge D. Group III metabotropic glutamate receptors: pharmacology, physiology and therapeutic potential. Neurochem Res 2014; 39:1876-94. [PMID: 25146900 DOI: 10.1007/s11064-014-1415-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/08/2014] [Accepted: 08/11/2014] [Indexed: 01/14/2023]
Abstract
Glutamate, the primary excitatory neurotransmitter in the central nervous system (CNS), exerts neuromodulatory actions via the activation of metabotropic glutamate (mGlu) receptors. There are eight known mGlu receptor subtypes (mGlu1-8), which are widely expressed throughout the brain, and are divided into three groups (I-III), based on signalling pathways and pharmacological profiles. Group III mGlu receptors (mGlu4/6/7/8) are primarily, although not exclusively, localised on presynaptic terminals, where they act as both auto- and hetero-receptors, inhibiting the release of neurotransmitter. Until recently, our understanding of the role of individual group III mGlu receptor subtypes was hindered by a lack of subtype-selective pharmacological tools. Recent advances in the development of both orthosteric and allosteric group III-targeting compounds, however, have prompted detailed investigations into the possible functional role of these receptors within the CNS, and revealed their involvement in a number of pathological conditions, such as epilepsy, anxiety and Parkinson's disease. The heterogeneous expression of group III mGlu receptor subtypes throughout the brain, as well as their distinct distribution at glutamatergic and GABAergic synapses, makes them ideal targets for therapeutic intervention. This review summarises the advances in subtype-selective pharmacology, and discusses the individual roles of group III mGlu receptors in physiology, and their potential involvement in disease.
Collapse
Affiliation(s)
- Marion S Mercier
- Centre for Synaptic Plasticity, School of Physiology and Pharmacology, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK,
| | | |
Collapse
|
31
|
Vadasz C, Saito M. New glutamatergic target for alcohol and substance use disorder medications. Psychopharmacology (Berl) 2014; 231:1429-31. [PMID: 24619644 DOI: 10.1007/s00213-014-3496-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 01/29/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Csaba Vadasz
- Nathan Kline Institute, Neurobehavioral Genetics, New York University School of Medicine, Psychiatry, New York, USA,
| | | |
Collapse
|
32
|
Zaniewska M, Przegaliński E, Filip M, Pilc A, Doller D. Inhibitory actions of mGlu4 receptor ligands on cocaine-, but not nicotine-, induced sensitizing and conditioning locomotor responses in rats. Pharmacol Rep 2014; 66:205-11. [PMID: 24911071 DOI: 10.1016/j.pharep.2013.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 12/06/2013] [Accepted: 12/09/2013] [Indexed: 10/25/2022]
Abstract
BACKGROUND Male Wistar rats were used to verify the hypothesis that metabotropic glutamate 4 (mGlu4) receptor ligands may modulate the locomotor effects evoked by cocaine or nicotine. METHODS The preferential mGlu4 receptor orthosteric agonist (2S)-2-amino-4-[hydroxy[hydroxy(4-hydroxy-3-methoxy-5-nitrophenyl)methyl]phosphoryl]butanoic acid (LSP1-2111) and the mGlu4 receptor positive allosteric modulator (+)-cis-N(1)-(3,4-dichlorophenyl)cyclohexane-1,2-dicarboxamide (Lu AF21934) were used in the study. Rats were given repeated pairings of a test environment with cocaine (10mg/kg), nicotine (0.4 mg/kg) or the respective vehicles for 5 days. On day 10, animals were challenged with cocaine (10mg/kg, cocaine sensitization), nicotine (0.4 mg/kg, nicotine sensitization) or vehicle (conditioned hyperlocomotion) in experimental cages. RESULTS Given on day 10, LSP1-2111 (3mg/kg) as well as Lu AF21934 (2.5-5mg/kg) decreased the expression of cocaine sensitization. In another set of experiments, LSP1-2111 (3mg/kg) and Lu AF21934 (5mg/kg) administered on day 10 attenuated the conditioned hyperlocomotion in rats treated repeatedly with cocaine. Neither LSP1-2111 (1-3mg/kg) nor Lu AF21934 (2.5-5mg/kg) changed the expression of nicotine sensitization and conditioned hyperlocomotion in rats treated repeatedly with nicotine. None of the mGlu4 receptor agonist/modulator altered the basal locomotor activity or acute hyperactivity to cocaine or nicotine. CONCLUSIONS The present data indicate that pharmacological stimulation of mGlu4 receptors reduces the cocaine-induced expression of sensitization as well as conditioned hyperactivity. In contrast, mGlu4 receptor activation seems to be devoid of any effect on the locomotor effects of nicotine.
Collapse
Affiliation(s)
- Magdalena Zaniewska
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland.
| | - Edmund Przegaliński
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Małgorzata Filip
- Laboratory of Drug Addiction Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Andrzej Pilc
- Department of Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Darío Doller
- Chemical and Pharmacokinetic Sciences, Lundbeck Research USA, Paramus, USA
| |
Collapse
|
33
|
Yan N, Chen N, Zhu H, Zhang J, Sim M, Ma Y, Wang W. High-frequency stimulation of nucleus accumbens changes in dopaminergic reward circuit. PLoS One 2013; 8:e79318. [PMID: 24244479 PMCID: PMC3828386 DOI: 10.1371/journal.pone.0079318] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 09/21/2013] [Indexed: 11/25/2022] Open
Abstract
Deep brain stimulation (DBS) of the nucleus accumbens (NAc) is a potential remedial therapy for drug craving and relapse, but the mechanism is poorly understood. We investigated changes in neurotransmitter levels during high frequency stimulation (HFS) of the unilateral NAc on morphine-induced rats. Sixty adult Wistar rats were randomized into five groups: the control group (administration of saline), the morphine-only group (systematic administration of morphine without electrode implantation), the morphine-sham-stimulation group (systematic administration of morphine with electrode implantation but not given stimulation), the morphine-stimulation group (systematic administration of morphine with electrode implantation and stimulation) and the saline-stimulation group (administration of saline with electrode implantation and stimulation). The stimulation electrode was stereotaxically implanted into the core of unilateral NAc and microdialysis probes were unilaterally lowered into the ipsilateral ventral tegmental area (VTA), NAc, and ventral pallidum (VP). Samples from microdialysis probes in the ipsilateral VTA, NAc, and VP were analyzed for glutamate (Glu) and γ-aminobutyric acid (GABA) by high-performance liquid chromatography (HPLC). The levels of Glu were increased in the ipsilateral NAc and VP of morphine-only group versus control group, whereas Glu levels were not significantly changed in the ipsilateral VTA. Furthermore, the levels of GABA decreased significantly in the ipsilateral NAc, VP, and VTA of morphine-only group when compared with control group. The profiles of increased Glu and reduced GABA in morphine-induced rats suggest that the presence of increased excitatory neurotransmission in these brain regions. The concentrations of the Glu significantly decreased while the levels of GABA increased in ipsilateral VTA, NAc, and VP in the morphine-stimulation group compared with the morphine-only group. No significant changes were seen in the morphine-sham stimulation group compared with the morphine-only group. These findings indicated that unilateral NAc stimulation inhibits the morphine-induced rats associated hyperactivation of excitatory neurotransmission in the mesocorticolimbic reward circuit.
Collapse
Affiliation(s)
- Na Yan
- Beijing Municipal Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
| | - Ning Chen
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Honghua Zhu
- Department of Gastroenterology, The Sixth People Hospital of Jinan, Jinan, China
| | - Jianguo Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Moira Sim
- Systems and Intervention Research Centre for Health, School of Medical Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Yu Ma
- Yuquan Hospital, Tsinghua University, Beijing, China
| | - Wei Wang
- Beijing Municipal Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
- Systems and Intervention Research Centre for Health, School of Medical Sciences, Edith Cowan University, Perth, Western Australia, Australia
| |
Collapse
|
34
|
Bahi A. Viral-mediated knockdown of mGluR7 in the nucleus accumbens mediates excessive alcohol drinking and increased ethanol-elicited conditioned place preference in rats. Neuropsychopharmacology 2013; 38:2109-19. [PMID: 22781839 PMCID: PMC3773660 DOI: 10.1038/npp.2012.122] [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] [Received: 01/30/2012] [Revised: 06/13/2012] [Accepted: 06/14/2012] [Indexed: 11/09/2022]
Abstract
Whether metabotropic glutamate 7 (mGluR7) -activation enhances or diminishes the reinforcing properties of psychostimulants remains unclear. We have previously shown that systemic mGluR7 activation reduced alcohol consumption and preference as well as locomotor-stimulating and rewarding properties of ethanol. In this study, we further examined the contribution of mGluR7 on the effect of ethanol within the nucleus accumbens (NAcc), a neural target for many drugs of abuse. Using short hairpin RNA (shRNA)-expressing lentiviral vectors (LV) to alter locally the activity of mGluR7 in male rats, we have shown that blocking mGluR7 expression increased ethanol consumption and preference in a two-bottle choice drinking paradigm with no effect either on saccharin or on quinine used for taste discrimination. In addition, mGluR7 knockdown increases preference for environments previously paired with low doses of ethanol in the conditioned place preference (CPP) test, as it shifted the dose-response curve for ethanol CPP to the left, indicating alterations in the rewarding effects of alcohol. More importantly, mGluR7 blockade in the dorsal striatum (DS) neither affected ethanol consumption nor ethanol-elicited CPP. These results show that levels of mGluR7 in the NAcc regulate responsiveness to alcohol. Taken together, these findings clearly demonstrate that mGluR7 signaling within the NAcc is a key modulator of functional responses to ethanol and offer an important target for regulating the addictive effects of alcohol.
Collapse
Affiliation(s)
- Amine Bahi
- Department of Anatomy, Faculty of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE,Department of Anatomy, Faculty of Medicine & Health Sciences, Tawam Medical Campus, United Arab Emirates University, PO Box 17666, Al Ain, UAE, Tel: +971 3 7137 516, Fax: +971 3 7672 033, E-mail:
| |
Collapse
|
35
|
Palazzo E, Marabese I, Luongo L, Boccella S, Bellini G, Giordano ME, Rossi F, Scafuro M, Novellis VD, Maione S. Effects of a metabotropic glutamate receptor subtype 7 negative allosteric modulator in the periaqueductal grey on pain responses and rostral ventromedial medulla cell activity in rat. Mol Pain 2013; 9:44. [PMID: 24004843 PMCID: PMC3846361 DOI: 10.1186/1744-8069-9-44] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 08/22/2013] [Indexed: 01/05/2023] Open
Abstract
The metabotropic glutamate receptor 7 (mGluR7) negative allosteric modulator, 6-(4-methoxyphenyl)-5-methyl-3-pyridin-4-ylisoxazolo[4,5-c]pyridin-4(5H)-one (MMPIP), was locally microinjected into the ventrolateral periaqueductal gray (VL PAG) and the effect on pain responses in formalin and spare nerve injury (SNI) -induced neuropathic pain models was monitored in the rat. The activity of rostral ventromedial medulla (RVM) “pronociceptive” ON and “antinociceptive” OFF cells was also evaluated. Intra–VL PAG MMPIP blocked the first and second phase of nocifensive behaviour in the formalin pain model. MMPIP increased the tail flick latency and simultaneously increased the activity of the OFF cells while inhibiting that of ON cells in rats with SNI of the sciatic nerve. MMPIP failed to modify nociceptive responses and associated RVM ON and OFF cell activity in sham rats. An increase in mGluR7 gene, protein and staining, the latter being associated with vesicular glutamate transporter-positive profiles, has been found in the VL PAG in SNI rats. Blockade of mGluR7 within the VL PAG has an antinociceptive effect in formalin and neuropathic pain models. VL PAG mGluR7 blockade offers a target for dis-inhibiting the VL PAG-RVM pathway and silencing pain in inflammatory and neuropathic pain models.
Collapse
Affiliation(s)
- Enza Palazzo
- Department of Anaesthesiology, Surgery and Emergency, The Second University of Naples, Piazza Luigi Miraglia 2, Naples 80178, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Yamasaki T, Kumata K, Yui J, Fujinaga M, Furutsuka K, Hatori A, Xie L, Ogawa M, Nengaki N, Kawamura K, Zhang MR. Synthesis and evaluation of [11C]MMPIP as a potential radioligand for imaging of metabotropic glutamate 7 receptor in the brain. EJNMMI Res 2013; 3:54. [PMID: 23870677 PMCID: PMC3751550 DOI: 10.1186/2191-219x-3-54] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 07/11/2013] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Metabotropic glutamate 7 (mGlu7) receptor is a crucial target protein for the development of pharmaceuticals against central nervous system disorders. In the present study, we synthesized [11C]MMPIP, a putative radioligand for mGlu7 (binding constant KB = 30 nM), and evaluated its potential for imaging of mGlu7 via in vitro and in vivo techniques. METHODS [11C]MMPIP was synthesized by the reaction of phenol precursor 3 with [11C]CH3I. In vitro autoradiography using [11C]MMPIP was performed on rat brain sections. To determine in vitro specific binding of [11C]MMPIP with mGlu7, a blocking study was conducted by co-incubation with excess AMN082, a selective antagonist for mGlu7, or unlabeled MMPIP. Positron emission tomography (PET) studies and ex vivo metabolite analysis were carried out on rat brains. RESULTS [11C]MMPIP was obtained with two specific activity (SA) levels of average 58 (conventional) and 3,800 (high SA) GBq/μmol, respectively. High radioactive signals derived from conventional [11C]MMPIP in the in vitro autoradiography were seen in the thalamus, medulla oblongata, and striatum, corresponding with comprehensive brain distributions of mGlu7. Co-incubation with ANM082 or unlabeled MMPIP reduced the radioactive signals in the brain sections, respectively. In the PET studies with [11C]MMPIP, no specific uptake relative to mGlu7 was found in the examined brain regions. CONCLUSION Despite in vitro specific binding of [11C]MMPIP with mGlu7, visualization of mGlu7 in the living brain using PET was not successful. Development of new ligand candidates with higher affinity for mGlu7 is necessary.
Collapse
Affiliation(s)
- Tomoteru Yamasaki
- Molecular Probe Program, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Cavas M, Scesa G, Navarro JF. Positive allosteric modulation of mGlu7 receptors by AMN082 affects sleep and wakefulness in the rat. Pharmacol Biochem Behav 2013; 103:756-63. [DOI: 10.1016/j.pbb.2012.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 12/02/2012] [Accepted: 12/12/2012] [Indexed: 11/30/2022]
|
38
|
Julio-Pieper M, O'Connor RM, Dinan TG, Cryan JF. Regulation of the brain-gut axis by group III metabotropic glutamate receptors. Eur J Pharmacol 2012; 698:19-30. [PMID: 23123053 DOI: 10.1016/j.ejphar.2012.10.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 10/11/2012] [Accepted: 10/22/2012] [Indexed: 01/14/2023]
Abstract
L-glutamate is produced by a great variety of peripheral tissues in both health and disease. Like other components of the glutamatergic system, metabotropic glutamate (mGlu) receptors also have a widespread distribution outside the central nervous system (CNS). In particular, group III mGlu receptors have been recently found in human stomach and colon revealing an extraordinary potential for these receptors in the treatment of peripheral disorders, including gastrointestinal dysfunction. The significance of these findings is that pharmacological tools originally designed for mGlu receptors in the CNS may also be directed towards new disease targets in the periphery. Targeting mGlu receptors can also be beneficial in the treatment of disorders involving central components together with gastrointestinal dysfunction, such as irritable bowel syndrome, which can be co-morbid with anxiety and depression. Conversely, the development of more specific therapeutic approaches for mGlu ligands both centrally as in the gut will depend on the elucidation of tissue-specific elements in mGlu receptor signalling.
Collapse
Affiliation(s)
- Marcela Julio-Pieper
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Av Universidad 330, Curauma, Valparaíso, Chile.
| | | | | | | |
Collapse
|
39
|
Hovelsø N, Sotty F, Montezinho LP, Pinheiro PS, Herrik KF, Mørk A. Therapeutic potential of metabotropic glutamate receptor modulators. Curr Neuropharmacol 2012; 10:12-48. [PMID: 22942876 PMCID: PMC3286844 DOI: 10.2174/157015912799362805] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2010] [Revised: 01/10/2011] [Accepted: 03/04/2011] [Indexed: 12/21/2022] Open
Abstract
Glutamate is the main excitatory neurotransmitter in the central nervous system (CNS) and is a major player in complex brain functions. Glutamatergic transmission is primarily mediated by ionotropic glutamate receptors, which include NMDA, AMPA and kainate receptors. However, glutamate exerts modulatory actions through a family of metabotropic G-protein-coupled glutamate receptors (mGluRs). Dysfunctions of glutamatergic neurotransmission have been implicated in the etiology of several diseases. Therefore, pharmacological modulation of ionotropic glutamate receptors has been widely investigated as a potential therapeutic strategy for the treatment of several disorders associated with glutamatergic dysfunction. However, blockade of ionotropic glutamate receptors might be accompanied by severe side effects due to their vital role in many important physiological functions. A different strategy aimed at pharmacologically interfering with mGluR function has recently gained interest. Many subtype selective agonists and antagonists have been identified and widely used in preclinical studies as an attempt to elucidate the role of specific mGluRs subtypes in glutamatergic transmission. These studies have allowed linkage between specific subtypes and various physiological functions and more importantly to pathological states. This article reviews the currently available knowledge regarding the therapeutic potential of targeting mGluRs in the treatment of several CNS disorders, including schizophrenia, addiction, major depressive disorder and anxiety, Fragile X Syndrome, Parkinson’s disease, Alzheimer’s disease and pain.
Collapse
Affiliation(s)
- N Hovelsø
- Department of Neurophysiology, H. Lundbeck A/S, Ottiliavej 9, 2500 Copenhagen-Valby, Denmark
| | | | | | | | | | | |
Collapse
|
40
|
Role for ventral pallidal GABAergic mechanisms in the regulation of ethanol self-administration. Psychopharmacology (Berl) 2012; 223:211-21. [PMID: 22552756 DOI: 10.1007/s00213-012-2709-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 03/27/2012] [Indexed: 11/27/2022]
Abstract
RATIONALE The striatopallidal medium spiny neurons have been viewed as a final common path for drug reward, and the ventral pallidum (VP) as a convergent point for hedonic and motivational signaling. The medium spiny neurons are GABAergic, but they colocalize enkephalin. OBJECTIVE The present study investigated the role of the GABAergic mechanisms of the VP in ethanol consumption. METHODS The effects of bilateral microinjections of GABA(A) and GABA(B) receptor agonists and antagonists into the VP on voluntary ethanol consumption were monitored in alcohol-preferring Alko alcohol rats given 90 min limited access to ethanol in their home cages every other day. The influences of coadministration of GABA and opioid receptor modulators were also studied. RESULTS The GABA(A) receptor agonist muscimol (1-10 ng/site) decreased ethanol intake dose-dependently, while administration of the GABA(A) receptor antagonist bicuculline (10-100 ng) had an opposite effect. The GABA(B) receptor agonist baclofen (3-30 ng) also suppressed ethanol intake, but the GABA(B) receptor antagonist saclofen (0.3-3 μg) failed to modify it. Animals coadministered with bicuculline (30 ng) and baclofen (30 ng) consumed ethanol significantly less than those treated with bicuculline alone. Coadministration of the μ-receptor agonist D-Ala2,N-Me-Phe4,Glyol5-enkephalin (DAMGO, 0.1 μg) with bicuculline counteracted, whereas the μ-receptor antagonist D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP, 1 μg) enhanced the bicuculline-induced increase of ethanol intake. When given alone, DAMGO decreased while CTOP increased ethanol intake. CONCLUSIONS The study provides evidence for the ventral pallidal GABAergic mechanisms participating in the regulation of ethanol consumption and supports earlier work suggesting a role for pallidal opioidergic transmission in ethanol reward.
Collapse
|
41
|
Li X, Higley A, Song R, Xi ZX. Effects of metabotropic glutamate receptor ligands on male sexual behavior in rats. Neuropharmacology 2012; 66:373-81. [PMID: 22968089 DOI: 10.1016/j.neuropharm.2012.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 08/01/2012] [Accepted: 08/13/2012] [Indexed: 01/08/2023]
Abstract
Metabotropic glutamate receptors (mGluRs), particularly mGluR2/3, mGluR5 and mGluR7, have received much attention in medication development for the treatment of drug addiction and other neuropsychiatric diseases. However, little is known as to whether mGluR ligands also alter natural sexual behavior, a possible unwanted effect when used in humans. In the present study, we used classical copulatory behaviors to evaluate the effects of LY379268 (a selective mGluR2/3 agonist), MPEP (a selective mGluR5 antagonist) and AMN082 (a selective mGluR7 agonist), on male sexual performance in rats. We found that systemic administration of LY379268 (1, 3 mg/kg, i.p.) had no effect, while MPEP (20 mg/kg, but not 10 mg/kg, i.p.) and AMN082 (10, 20 mg/kg, but not 3 mg/kg) produced a significant and dose-dependent reduction in both sex-seeking and sex-performance behaviors, manifested as an increase in mount or intromission latency and time required for ejaculation, and a reduction in mount or intromission frequency. This inhibition lasted for about 30-60 min. These findings suggest that compounds that target mGluR5 or mGluR7, but not mGluR2/3, may have short-term inhibitory effects on male sexual performance. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.
Collapse
Affiliation(s)
- Xia Li
- National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD 21224, USA
| | | | | | | |
Collapse
|
42
|
Flor PJ, Acher FC. Orthosteric versus allosteric GPCR activation: the great challenge of group-III mGluRs. Biochem Pharmacol 2012; 84:414-24. [PMID: 22554564 DOI: 10.1016/j.bcp.2012.04.013] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 04/17/2012] [Accepted: 04/17/2012] [Indexed: 10/28/2022]
Abstract
Group-III metabotropic glutamate receptors (mGluRs) comprise four structurally related brain and retinal G protein-coupled receptors (GPCRs), mGluR4, mGluR6, mGluR7 and mGluR8, which receive much attention as promising targets for nervous system drugs. In particular, activation of mGluR4 is a major focus for the development of new therapeutics in Parkinson's disease, while mGluR7 activation is considered a potential approach for future treatments of specific psychiatric conditions. The first generation group-III mGluR agonists, e.g.l-AP4 and l-SOP, are characterized by an essential phosphonate functional group, which became a major limitation for the development of systemically active, potent and receptor subtype-selective drugs. Recently however, two approaches emerged in parallel providing resolution to this constraint: in silico high-throughput screening of chemical libraries against a 3D-model of the mGluR4 extracellular domain identified a hit that was optimized into a series of potent and subtype-selective orthosteric agonists with drug-like properties and novel chemotype structures; secondly, high-throughput random screening of chemical libraries against recombinantly expressed group-III receptors identified diverse chemical sets of allosteric agonists and positive modulators, which are drug-like, display selectivity for mGluR4, mGluR7, or mGluR8 and act via novel pharmacological sites. Here, we illustrate new scientific insights obtained via the use of those strategies. Also, we compare advantages and disadvantages of both approaches to identify the desired group-III mGluR activators and we conclude with suggestions how to employ those discovery strategies with success for the identification, optimization, and development of clinical drug candidates; this may have important implications for the entire field of GPCR research.
Collapse
Affiliation(s)
- Peter J Flor
- Faculty of Biology and Preclinical Medicine, University of Regensburg, Regensburg, Germany.
| | | |
Collapse
|
43
|
Li X, Xi ZX, Markou A. Metabotropic glutamate 7 (mGlu7) receptor: a target for medication development for the treatment of cocaine dependence. Neuropharmacology 2012; 66:12-23. [PMID: 22546614 DOI: 10.1016/j.neuropharm.2012.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Revised: 03/21/2012] [Accepted: 04/11/2012] [Indexed: 11/19/2022]
Abstract
Brain glutamate has been shown to play an important role in reinstatement to drug seeking, a behavior considered to be of relevance to relapse to drug taking in humans. Therefore, glutamate receptors, in particular metabotropic glutamate (mGlu) receptors, have become important targets for medication development for the treatment of drug dependence. In this review article, we focus on the mGlu7 receptor subtype, and discuss recent findings with AMN082, a selective mGlu7 receptor allosteric agonist, in animal models with relevance to drug dependence. Systemic or local administration of AMN082 into the nucleus accumbens (NAc), a critical brain region involved in reward and drug dependence processes, inhibited the reinforcing and motivational effects of cocaine, heroin and ethanol, as assessed by the intravenous drug self-administration procedure. In addition, AMN082 inhibited the reward-enhancing effects induced by cocaine, as assessed in the intracranial self-stimulation procedure, and cocaine- or cue-induced reinstatement of drug-seeking behavior. In vivo microdialysis studies indicated that systemic or intra-NAc administration of AMN082 significantly decreased extracellular γ-aminobutyric acid (GABA) and elevated extracellular glutamate, but had no effect on extracellular dopamine in the NAc, suggesting that a non-dopaminergic mechanism underlies the effects of AMN082 on the actions of cocaine. Further, data indicated that AMN082-induced changes in glutamate were the net effect of two actions: one is the direct inhibition of glutamate release by activation of mGlu7 receptors on glutamatergic neurons; another is the indirect increases of glutamate release mediated by decreases in GABA transmission. These increases in extracellular glutamate functionally antagonized cocaine-induced inhibition of NAc-ventral pallidum GABAergic neurotransmission, and therefore, the rewarding effects of cocaine. In addition, elevated extracellular glutamate activated presynaptic mGlu2/3 autoreceptors which in turn inhibited cocaine priming- or cue-induced enhancement of glutamate release and reinstatement of drug-seeking behavior. Taken together, these findings suggest that the mGlu7 receptor is an important target for medication development for the treatment of drug dependence. AMN082 or other mGlu7 receptor allosteric agonists may have potential as novel pharmacotherapies for cocaine addiction. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.
Collapse
Affiliation(s)
- Xia Li
- Department of Psychiatry, M/C 0603, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0603, USA.
| | | | | |
Collapse
|
44
|
Bahi A, Fizia K, Dietz M, Gasparini F, Flor PJ. Pharmacological modulation of mGluR7 with AMN082 and MMPIP exerts specific influences on alcohol consumption and preference in rats. Addict Biol 2012; 17:235-47. [PMID: 21392179 DOI: 10.1111/j.1369-1600.2010.00310.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Growing evidence supports a role for the central nervous system (CNS) neurotransmitter L-glutamate and its metabotropic receptors (mGluRs) in drug addiction in general and alcohol-use disorders in particular. Alcohol dependence, for instance, has a genetic component, and the recent discovery that variations in the gene coding for mGluR7 modulate alcohol consumption further validates involvement of the L-glutamate system. Consequently, increasing interest emerges in developing L-glutamatergic therapies for the treatment of alcohol abuse and dependence. To this end, we performed a detailed behavioral pharmacology study to investigate the regulation of alcohol consumption and preference following administration of the mGluR7-selective drugs N,N'-dibenzyhydryl-ethane-1,2-diamine dihydrochloride (AMN082) and 6-(4-Methoxyphenyl)-5-methyl-3-(4-pyridinyl)-isoxazolo[4,5-c]pyridin-4(5H)-one hydrochloride (MMPIP). Upon administration of the allosteric agonist AMN082 (10 mg/kg, i.p.) in rats, there was a significant decrease in ethanol consumption and preference, without affecting ethanol blood metabolism. In contrast, mGluR7 blockade with MMPIP (10 mg/kg, i.p.) showed an increase in alcohol intake and reversed AMN082's effect on ethanol consumption and preference. Both mGluR7-directed pharmacological tools had no effect on total fluid intake, taste preference, or on spontaneous locomotor activity. In conclusion, these findings support a specific regulatory role for mGluR7 on alcohol drinking and preference and provide evidence for the use of AMN082-type drugs as potential new treatments for alcohol-use disorders in man.
Collapse
Affiliation(s)
- Amine Bahi
- Faculty of Biology and Preclinical Medicine, Laboratory of Molecular & Cellular Neurobiology, University of Regensburg, Universitätsstraße 31, Regensburg, Germany.
| | | | | | | | | |
Collapse
|
45
|
Cleva RM, Olive MF. Metabotropic glutamate receptors and drug addiction. ACTA ACUST UNITED AC 2012; 1:281-295. [DOI: 10.1002/wmts.18] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
46
|
The selective metabotropic glutamate receptor 7 allosteric agonist AMN082 prevents reinstatement of extinguished ethanol-induced conditioned place preference in mice. Pharmacol Biochem Behav 2012; 101:193-200. [PMID: 22269296 DOI: 10.1016/j.pbb.2012.01.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Revised: 01/07/2012] [Accepted: 01/09/2012] [Indexed: 11/20/2022]
Abstract
Alcohol dependence is considered a major public health problem in modern societies. The role for glutamatergic neurotransmission in the reinforcing effects of ethanol is becoming increasingly evident. Our previous findings have shown that in rats, the mGluR7 positive allosteric agonist AMN082, but not its allosteric antagonist MMPIP, prevented ethanol consumption and preference in the two-bottle choice paradigm. This study was conducted to determine the effects of AMN082 and MMPIP on the extinction and reinstatement of ethanol-elicited place preference (CPP) in C57BL/6 mice. AMN082 and MMPIP were administered during extinction of ethanol CPP to determine whether mGluR7 signaling is required. Furthermore, the effects of AMN082 and MMPIP on reinstatement of CPP were also evaluated. Finally, spontaneous locomotor activity and ethanol pharmacokinetics were assessed following systemic administration of AMN082 and MMPIP. Our results indicate that mGluR7 pharmacological modulation had no effect on ethanol-elicited CPP extinction. In contrast, mGluR7 activation using AMN082 reduced ethanol-induced CPP reinstatement, an effect reversed by co-administration of MMPIP. Collectively, these results indicate, for the first time, that activation of the mGluR7 receptor is effective in reducing the reinstatement of conditioned rewarding effects of ethanol. Taken together, the efficacy of AMN082 on the various phases of alcohol-CPP could represent an interesting pharmacological approach and could open a new line of research for the development of therapies to reduce ethanol intake in patients.
Collapse
|
47
|
Srivastava R, Xi ZX, Gardner EL. Cocaine-taking and cocaine-seeking behaviors in rats remain stable after systemic administration of GYKI 52466: a non-competitive AMPA receptor antagonist. Neurosci Lett 2011; 508:106-9. [PMID: 22206835 DOI: 10.1016/j.neulet.2011.12.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 12/01/2011] [Accepted: 12/11/2011] [Indexed: 01/11/2023]
Abstract
Given the posited role of enhanced AMPA-mediated synaptic transmission in relapse to drug seeking, we investigated whether systemic administration of the AMPA receptor antagonist GYKI 52466 inhibits cocaine-taking and cocaine-seeking behavior in rats. Rats were trained to self-administer cocaine until stable self-administration was achieved. Effects of GYKI 52466 (1, 3, or 10mg/kg, i.v.) on cocaine self-administration were assessed. Animals were allowed to re-establish stable cocaine self-administration and were then behaviorally extinguished from drug taking. The effects of GYKI 52466 (3, 10mg/kg, i.v.) on cocaine-induced reinstatement of drug-seeking behavior were assessed. We found that GYKI 52466 failed to inhibit cocaine-taking and cocaine-seeking in both the self-administration and reinstatement paradigms. We suggest that although AMPA receptors may be involved in cocaine reward and addiction, the AMPA receptor antagonist GYKI 52466 has low therapeutic potential for cocaine addiction treatment.
Collapse
Affiliation(s)
- Ratika Srivastava
- National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD 21224, USA.
| | | | | |
Collapse
|
48
|
Bahi A. RETRACTED: The pre-synaptic metabotropic glutamate receptor 7 “mGluR7” is a critical modulator of ethanol sensitivity in mice. Neuroscience 2011; 199:13-23. [DOI: 10.1016/j.neuroscience.2011.10.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 10/16/2011] [Accepted: 10/17/2011] [Indexed: 10/15/2022]
|
49
|
Root DH, Fabbricatore AT, Pawlak AP, Barker DJ, Ma S, West MO. Slow phasic and tonic activity of ventral pallidal neurons during cocaine self-administration. Synapse 2011; 66:106-27. [PMID: 21953543 DOI: 10.1002/syn.20990] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 09/14/2011] [Indexed: 02/06/2023]
Abstract
Ventral pallidal (VP) neurons exhibit rapid phasic firing patterns within seconds of cocaine-reinforced responses. The present investigation examined whether VP neurons exhibited firing rate changes: (1) over minutes during the inter-infusion interval (slow phasic patterns) and/or (2) over the course of the several-hour self-administration session (tonic firing patterns) relative to pre-session firing. Approximately three-quarters (43/54) of VP neurons exhibited slow phasic firing patterns. The most common pattern was a post-infusion decrease in firing followed by a progressive reversal of firing over minutes (51.16%; 22/43). Early reversals were predominantly observed anteriorly whereas progressive and late reversals were observed more posteriorly. Approximately half (51.85%; 28/54) of the neurons exhibited tonic firing patterns consisting of at least a two-fold change in firing. Most cells decreased firing during drug loading, remained low over self-administration maintenance, and reversed following lever removal. Over a whole experiment (tonic) timescale, the majority of neurons exhibited an inverse relationship between calculated drug level and firing rates during loading and post-self-administration behaviors. Fewer neurons exhibited an inverse relationship of calculated drug level and tonic firing rate during self-administration maintenance but, among those that did, nearly all were progressive reversal neurons. The present results show that, similar to its main afferent the nucleus accumbens, VP exhibits both slow phasic and tonic firing patterns during cocaine self-administration. Given that VP neurons are principally GABAergic, the predominant slow phasic decrease and tonic decrease firing patterns within the VP may indicate a disinhibitory influence upon its thalamocortical, mesolimbic, and nigrostriatal targets during cocaine self-administration.
Collapse
Affiliation(s)
- David H Root
- Department of Psychology, Rutgers University, New Brunswick, New Jersey 08903, USA
| | | | | | | | | | | |
Collapse
|
50
|
Enhanced GABAergic tone in the ventral pallidum: memory of unpleasant experiences? Neuroscience 2011; 196:131-46. [DOI: 10.1016/j.neuroscience.2011.08.058] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 08/10/2011] [Accepted: 08/25/2011] [Indexed: 01/08/2023]
|