1
|
Lu G, Ma F, Wei P, Ma M, Tran VNH, Baldo BA, Li L. Cocaine-Induced Remodeling of the Rat Brain Peptidome: Quantitative Mass Spectrometry Reveals Anatomically Specific Patterns of Cocaine-Regulated Peptide Changes. ACS Chem Neurosci 2025; 16:128-140. [PMID: 39810605 PMCID: PMC11736046 DOI: 10.1021/acschemneuro.4c00327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025] Open
Abstract
Addiction to psychostimulants, including cocaine, causes widespread morbidity and mortality and is a major threat to global public health. Currently, no pharmacotherapies can successfully treat psychostimulant addiction. The neuroactive effects of cocaine and other psychostimulants have been studied extensively with respect to their modulation of monoamine systems (particularly dopamine); effects on neuropeptide systems have received less attention. Here, we employed mass spectrometry (MS) methods to characterize cocaine-induced peptidomic changes in the rat brain. Label-free peptidomic analysis using liquid chromatography coupled with tandem MS (LC-MS/MS) was used to describe the dynamic changes of endogenous peptides in five brain regions (nucleus accumbens, dorsal striatum, prefrontal cortex, amygdala, and hypothalamus) following an acute systemic cocaine challenge. The improved sensitivity and specificity of this method, coupled with quantitative assessment, enabled the identification of 1376 peptides derived from 89 protein precursors. Our data reveal marked, region-specific changes in peptide levels in the brain induced by acute cocaine exposure, with peptides in the cholecystokinin and melanin-concentrating hormone families being significantly affected. These findings offer new insights into the region-specific effects of cocaine and could pave the way for developing new therapies to treat substance use disorders and related psychiatric conditions.
Collapse
Affiliation(s)
- Gaoyuan Lu
- School of Pharmacy, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Fengfei Ma
- School of Pharmacy, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Pingli Wei
- Department of Chemistry, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
| | - Min Ma
- School of Pharmacy, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Vu Ngoc Huong Tran
- School of Pharmacy, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Brian A Baldo
- Department of Psychiatry, University of Wisconsin─Madison, Madison, Wisconsin 53719, United States
- Neuroscience Training Program, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
- Department of Chemistry, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
- Lachman Institute for Pharmaceutical Development, School of Pharmacy, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin─Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
2
|
Abstract
The development of nonpeptide systemically active small-molecule NOP-targeted ligands has contributed tremendously to validating the NOP receptor as a promising target for therapeutics. Although a NOP-targeted compound is not yet approved for clinical use, a few NOP ligands are in clinical trials for various indications. Both successful and failed human clinical trials with NOP ligands provide opportunities for rational development of new and improved NOP-targeted compounds. A few years after the discovery of the NOP receptor in 1994, and its de-orphanization upon discovery of the endogenous peptide nociceptin/orphanin FQ (N/OFQ) in 1995, there was a significant effort in the pharmaceutical industry to discover nonpeptide NOP ligands from hits obtained from high-throughput screening campaigns of compound libraries. Depending on the therapeutic indication to be pursued, NOP agonists and antagonists were discovered, and some were optimized as clinical candidates. Advances such as G protein-coupled receptor (GPCR) structure elucidation, functional selectivity in ligand-driven GPCR activation, and multi-targeted ligands provide new scope for the rational design of novel NOP ligands fine-tuned for successful clinical translation. This article reviews the field of nonpeptide NOP ligand drug design in the context of these exciting developments and highlights new optimized nonpeptide NOP ligands possessing interesting functional profiles, which are particularly attractive for several unmet clinical applications involving NOP receptor pharmacomodulation.
Collapse
|
3
|
Alcaraz-Iborra M, Navarrete F, Rodríguez-Ortega E, de la Fuente L, Manzanares J, Cubero I. Different Molecular/Behavioral Endophenotypes in C57BL/6J Mice Predict the Impact of OX 1 Receptor Blockade on Binge-Like Ethanol Intake. Front Behav Neurosci 2017; 11:186. [PMID: 29066961 PMCID: PMC5641301 DOI: 10.3389/fnbeh.2017.00186] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 09/25/2017] [Indexed: 12/16/2022] Open
Abstract
Ethanol (EtOH) research has focused on stages of dependence. It is of paramount importance to more deeply understand the neurobehavioral factors promoting increased risk for EtOH binge drinking during the early stages of the addiction cycle. The first objective of this study was to evaluate whether C57BL/6J mice showing high drinking in the dark (DID) exhibit neurobehavioral traits known to contribute to EtOH binge-drinking disorders. Comparing high vs. low drinkers (HD/LD), we evaluated different types of basal anxiety-like responses, EtOH preference and sensitivity to the reinforcing properties of EtOH, and basal mRNA expression of the OX1/OX2 receptors (OX1r/OX2r) within the prefrontal cortex (PFC) and the nucleus accumbens (NAcc). Additionally, we tested binge drinking by LD/HD in response to a selective OX1r antagonist following intermittent episodes of DID (iDID). We report that DID consistently segregates two neurobehavioral endophenotypes, HD vs. LD, showing differences in neophobia and/or impulsivity/compulsivity traits. Additionally, HD mice show decreased basal OX1r and OX2r mRNA expression within the NAcc and elevated OX1r within the PFC. Exposure to several intermittent episodes of EtOH DID triggered a rapid increase in EtOH intake over time in LD mice matching that observed in HD mice. Despite HD/LD endophenotypes did not show differences in EtOH intake, they still predicted the response to a pharmacological challenge with a selective OX1r antagonist. The present data underscore the relevance of HD/LD endophenotypes stemming from DID procedures for exploring neurobehavioral processes underlying the early stages of the addiction cycle and EtOH binge-drinking disorders.
Collapse
Affiliation(s)
- Manuel Alcaraz-Iborra
- Laboratorio de Psicobiología, Departamento de Psicologia, Universidad de Almería, Almería, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Miguel Hernández de Elche, Elche, Spain
| | - Elisa Rodríguez-Ortega
- Laboratorio de Psicobiología, Departamento de Psicologia, Universidad de Almería, Almería, Spain
| | - Leticia de la Fuente
- Laboratorio de Psicobiología, Departamento de Psicologia, Universidad de Almería, Almería, Spain
| | - Jorge Manzanares
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Miguel Hernández de Elche, Elche, Spain
| | - Inmaculada Cubero
- Laboratorio de Psicobiología, Departamento de Psicologia, Universidad de Almería, Almería, Spain.,Institute of Biomedical Sciences, Universidad Autonoma de Chile, Santiago de Chile, Chile
| |
Collapse
|
4
|
Ruzza C, Calò G, Di Maro S, Pacifico S, Trapella C, Salvadori S, Preti D, Guerrini R. Neuropeptide S receptor ligands: a patent review (2005-2016). Expert Opin Ther Pat 2016; 27:347-362. [PMID: 27788040 DOI: 10.1080/13543776.2017.1254195] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Neuropeptide S (NPS) is a 20-residue peptide and endogenous ligand of the NPS receptor (NPSR). This receptor was a formerly orphan GPCR whose activation increases calcium and cyclic adenosine monophosphate levels. The NPS/NPSR system is expressed in several brain regions where it controls important biological functions including locomotor activity, arousal and sleep, anxiety, food intake, memory, pain, and drug addiction. Areas covered: This review furnishes an updated overview of the patent literature covering NPSR ligands since 2005, when the first example of an NPSR antagonist was disclosed. Expert opinion: Several potent NPSR antagonists are available as valuable pharmacological tools despite showing suboptimal pharmacokinetic properties in vivo. The optimization of these ligands is needed to speed up their potential clinical advancement as pharmaceuticals to treat drug addiction. In order to support the design of novel NPSR antagonists, we performed a ligand-based conformational analysis recognizing some structural requirements for NPSR antagonism. The identification of small-molecule NPSR agonists now represents an unmet challenge to be addressed. These molecules will allow investigation of the beneficial effects of selective NPSR activation in a large panel of psychiatric disorders and to foresee their therapeutic potential as anxiolytics, nootropics, and analgesics.
Collapse
Affiliation(s)
- Chiara Ruzza
- a Department of Medical Sciences, Section of Pharmacology, School of Medicine and National Institute of Neuroscience , University of Ferrara , Ferrara , Italy
| | - Girolamo Calò
- a Department of Medical Sciences, Section of Pharmacology, School of Medicine and National Institute of Neuroscience , University of Ferrara , Ferrara , Italy
| | | | - Salvatore Pacifico
- c Department of Chemical and Pharmaceutical Sciences , University of Ferrara , Ferrara , Italy
| | - Claudio Trapella
- c Department of Chemical and Pharmaceutical Sciences , University of Ferrara , Ferrara , Italy
| | - Severo Salvadori
- c Department of Chemical and Pharmaceutical Sciences , University of Ferrara , Ferrara , Italy
| | - Delia Preti
- c Department of Chemical and Pharmaceutical Sciences , University of Ferrara , Ferrara , Italy
| | - Remo Guerrini
- c Department of Chemical and Pharmaceutical Sciences , University of Ferrara , Ferrara , Italy
| |
Collapse
|
5
|
Miranda-Morales RS, Pautassi RM. Pharmacological characterization of the nociceptin/orphanin FQ receptor on ethanol-mediated motivational effects in infant and adolescent rats. Behav Brain Res 2016; 298:88-96. [PMID: 25907741 DOI: 10.1016/j.bbr.2015.04.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 04/09/2015] [Accepted: 04/10/2015] [Indexed: 01/17/2023]
Abstract
Activation of nociceptin/orphanin FQ (NOP) receptors attenuates ethanol drinking and prevents relapse in adult rodents. In younger rodents (i.e., infant rats), activation of NOP receptors blocks ethanol-induced locomotor activation but does not attenuate ethanol intake. The aim of the present study was to extend the analysis of NOP modulation of ethanol's effects during early ontogeny. Aversive and anxiolytic effects of ethanol were measured in infant and adolescent rats via conditioned taste aversion and the light-dark box test; whereas ethanol-induced locomotor activity and ethanol intake was measured in adolescents only. Before these tests, infant rats were treated with the natural ligand of NOP receptors, nociceptin (0.0, 0.5 or 1.0 μg) and adolescent rats were treated with the specific agonist Ro 64-6198 (0.0, 0.1 or 0.3 mg/kg). The activation of NOP receptors attenuated ethanol-induced anxiolysis in adolescents only, and had no effect on ethanol's aversive effects. Administration of Ro 64-6198 blocked ethanol-induced locomotor activation but did not modify ethanol intake patterns. The attenuation of ethanol stimulating and anxiolytic effect by activation of NOP receptors indicates a modulatory role of this receptor on ethanol effects, which is expressed early in ontogeny.
Collapse
Affiliation(s)
- Roberto Sebastián Miranda-Morales
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), 5016 Córdoba, Argentina.
| | - Ricardo M Pautassi
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), 5016 Córdoba, Argentina; Facultad de Psicología, Universidad Nacional de Córdoba, 5000 Córdoba, Argentina
| |
Collapse
|
6
|
Ayanwuyi LO, Stopponi S, Ubaldi M, Cippitelli A, Nasuti C, Damadzic R, Heilig M, Schank J, Cheng K, Rice KC, Ciccocioppo R. Neurokinin 1 receptor blockade in the medial amygdala attenuates alcohol drinking in rats with innate anxiety but not in Wistar rats. Br J Pharmacol 2015; 172:5136-46. [PMID: 26275374 DOI: 10.1111/bph.13280] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 07/27/2015] [Accepted: 08/02/2015] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND PURPOSE Substance P and its preferred neurokinin receptor NK1 have been implicated in stress and anxiety and have been proposed as possible therapeutic targets for the treatment of anxiety/depression. Attention is also being focused on the role this neuropeptide system may play in drug addiction, because stress-related mechanisms promote drug abuse. EXPERIMENTAL APPROACH The effects of the rat-specific NK1 receptor antagonist, L822429, on alcohol intake and seeking behaviour was investigated in genetically selected Marchigian Sardinian alcohol preferring rats. These rats demonstrate an anxious phenotype and are highly sensitive to stress and stress-induced drinking. KEY RESULTS Systemic administration of L822429 significantly reduced operant alcohol self-administration in Marchigian Sardinian alcohol preferring rats, but did not reduce alcohol self-administration in stock Wistar rats. NK1 receptor antagonism also attenuated yohimbine-induced reinstatement of alcohol seeking at all doses tested but had no effect on cue-induced reinstatement of alcohol seeking. L822429 reduced operant alcohol self-administration when injected into the lateral cerebroventricles or the medial amygdala. L822429 injected into the medial amygdala also significantly reduced anxiety-like behaviour in the elevated plus maze test. No effects on alcohol intake were observed following injection of L822429 into the dorsal or the ventral hippocampus. Conclusions and Implications Our results suggest that NK1 receptor antagonists may be useful for the treatment of alcohol addiction associated with stress or comorbid anxiety disorders. The medial amygdala appears to be an important brain site of action of NK1 receptor antagonism.
Collapse
Affiliation(s)
- Lydia O Ayanwuyi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, 62032, Italy
| | - Serena Stopponi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, 62032, Italy
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, 62032, Italy
| | - Andrea Cippitelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, 62032, Italy
| | - Cinzia Nasuti
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, 62032, Italy
| | - Ruslan Damadzic
- Laboratory of Clinical and Translational Studies, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD, 20892-1108, USA
| | - Markus Heilig
- Laboratory of Clinical and Translational Studies, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD, 20892-1108, USA
| | - Jesse Schank
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Kejun Cheng
- Drug Design and Synthesis Section, Chemical Biology Research Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Kenner C Rice
- Drug Design and Synthesis Section, Chemical Biology Research Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, 62032, Italy
| |
Collapse
|
7
|
Aujla H, Nedjadrasul D. Low-dose Nociceptin/Orphanin FQ reduces anxiety-like performance in alcohol-withdrawn, but not alcohol-naïve, Male Wistar rats. Neuropharmacology 2015; 93:1-6. [PMID: 25617519 DOI: 10.1016/j.neuropharm.2015.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 01/08/2015] [Accepted: 01/10/2015] [Indexed: 10/24/2022]
Abstract
Alcohol withdrawal is associated with neuroadaptation of stress-regulatory systems, including transmission of neuropeptides that have been implicated in anxiety-like performance. Nociceptin/Orphanin FQ (N/OFQ), an endogenous neuropeptide ligand at the NOP receptor, has been implicated in stress and has previously been shown to attenuate or exacerbate anxiety-like performance in rats following a biphasic dose response function. In addition, divergent actions on anxiety-like performance have been observed in alcohol-withdrawn vs. control animals, suggesting alcohol-induced alteration of N/OFQ transmission. In order to differentiate between whether this divergence resulted from a "switch" in the actions of N/OFQ vs. increased sensitivity in N/OFQ transmission, we assessed the actions of low doses of N/OFQ (0, 0.125, 0.25, or 0.5 μg) on two tests of anxiety, the shock-probe defensive burying and elevated plus maze tests, three weeks after the termination of a six-day regimen of alcohol or vehicle administration via intragastric intubation. Consistent with increased sensitivity in N/OFQ resulting from a history of alcohol intake, administration of a low dose of N/OFQ (0.25 μg) selectively attenuated anxiety-like behaviors in animals with a history of alcohol intake while controls did not exhibit any changes in performance. The present results suggest that withdrawal from alcohol produces an enduring increase in sensitivity in N/OFQ transmission - a finding that is consistent with previous studies demonstrating altered transmission in related neuropeptide systems.
Collapse
Affiliation(s)
- Harinder Aujla
- Department of Psychology, University of Winnipeg, 515 Portage Ave, R3B 2E9 Winnipeg, Canada.
| | - Daniel Nedjadrasul
- Department of Psychology, University of Winnipeg, 515 Portage Ave, R3B 2E9 Winnipeg, Canada
| |
Collapse
|
8
|
Lohman RJ, Harrison RS, Ruiz-Gómez G, Hoang HN, Shepherd NE, Chow S, Hill TA, Madala PK, Fairlie DP. Helix-constrained nociceptin peptides are potent agonists and antagonists of ORL-1 and nociception. VITAMINS AND HORMONES 2015; 97:1-55. [PMID: 25677767 DOI: 10.1016/bs.vh.2014.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nociceptin (orphanin FQ) is a 17-residue neuropeptide hormone with roles in both nociception and analgesia. It is an opioid-like peptide that binds to and activates the G-protein-coupled receptor opioid receptor-like-1 (ORL-1, NOP, orphanin FQ receptor, kappa-type 3 opioid receptor) on central and peripheral nervous tissue, without activating classic delta-, kappa-, or mu-opioid receptors or being inhibited by the classic opioid antagonist naloxone. The three-dimensional structure of ORL-1 was recently published, and the activation mechanism is believed to involve capture by ORL-1 of the high-affinity binding, prohelical C-terminus. This likely anchors the receptor-activating N-terminus of nociception nearby for insertion in the membrane-spanning helices of ORL-1. In search of higher agonist potency, two lysine and two aspartate residues were strategically incorporated into the receptor-binding C-terminus of the nociceptin sequence and two Lys(i)→Asp(i+4) side chain-side chain condensations were used to generate lactam cross-links that constrained nociceptin into a highly stable α-helix in water. A cell-based assay was developed using natively expressed ORL-1 receptors on mouse neuroblastoma cells to measure phosphorylated ERK as a reporter of agonist-induced receptor activation and intracellular signaling. Agonist activity was increased up to 20-fold over native nociceptin using a combination of this helix-inducing strategy and other amino acid modifications. An NMR-derived three-dimensional solution structure is described for a potent ORL-1 agonist derived from nociceptin, along with structure-activity relationships leading to the most potent known α-helical ORL-1 agonist (EC₅₀ 40 pM, pERK, Neuro-2a cells) and antagonist (IC₅₀ 7 nM, pERK, Neuro-2a cells). These α-helix-constrained mimetics of nociceptin(1-17) had enhanced serum stability relative to unconstrained peptide analogues and nociceptin itself, were not cytotoxic, and displayed potent thermal analgesic and antianalgesic properties in rats (ED₅₀ 70 pmol, IC₅₀ 10 nmol, s.c.), suggesting promising uses in vivo for the treatment of pain and other ORL-1-mediated responses.
Collapse
Affiliation(s)
- Rink-Jan Lohman
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Rosemary S Harrison
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Gloria Ruiz-Gómez
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Huy N Hoang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Nicholas E Shepherd
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Shiao Chow
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Timothy A Hill
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Praveen K Madala
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - David P Fairlie
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
9
|
Lang R, Gundlach AL, Holmes FE, Hobson SA, Wynick D, Hökfelt T, Kofler B. Physiology, signaling, and pharmacology of galanin peptides and receptors: three decades of emerging diversity. Pharmacol Rev 2015; 67:118-75. [PMID: 25428932 DOI: 10.1124/pr.112.006536] [Citation(s) in RCA: 238] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Galanin was first identified 30 years ago as a "classic neuropeptide," with actions primarily as a modulator of neurotransmission in the brain and peripheral nervous system. Other structurally-related peptides-galanin-like peptide and alarin-with diverse biologic actions in brain and other tissues have since been identified, although, unlike galanin, their cognate receptors are currently unknown. Over the last two decades, in addition to many neuronal actions, a number of nonneuronal actions of galanin and other galanin family peptides have been described. These include actions associated with neural stem cells, nonneuronal cells in the brain such as glia, endocrine functions, effects on metabolism, energy homeostasis, and paracrine effects in bone. Substantial new data also indicate an emerging role for galanin in innate immunity, inflammation, and cancer. Galanin has been shown to regulate its numerous physiologic and pathophysiological processes through interactions with three G protein-coupled receptors, GAL1, GAL2, and GAL3, and signaling via multiple transduction pathways, including inhibition of cAMP/PKA (GAL1, GAL3) and stimulation of phospholipase C (GAL2). In this review, we emphasize the importance of novel galanin receptor-specific agonists and antagonists. Also, other approaches, including new transgenic mouse lines (such as a recently characterized GAL3 knockout mouse) represent, in combination with viral-based techniques, critical tools required to better evaluate galanin system physiology. These in turn will help identify potential targets of the galanin/galanin-receptor systems in a diverse range of human diseases, including pain, mood disorders, epilepsy, neurodegenerative conditions, diabetes, and cancer.
Collapse
Affiliation(s)
- Roland Lang
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| | - Andrew L Gundlach
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| | - Fiona E Holmes
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| | - Sally A Hobson
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| | - David Wynick
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| | - Tomas Hökfelt
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| | - Barbara Kofler
- Department of Dermatology (R.L.) and Laura Bassi Centre of Expertise, Department of Pediatrics (B.K.), Paracelsus Private Medical University, Salzburg, Austria; The Florey Institute of Neuroscience and Mental Health, and Florey Department of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia (A.L.G.); Schools of Physiology and Pharmacology and Clinical Sciences, Bristol University, Bristol, United Kingdom (F.E.H., S.A.H., D.W.); and Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (T.H.)
| |
Collapse
|
10
|
Meinhardt MW, Sommer WH. Postdependent state in rats as a model for medication development in alcoholism. Addict Biol 2015; 20:1-21. [PMID: 25403107 DOI: 10.1111/adb.12187] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Rational development of novel therapeutic strategies for alcoholism requires understanding of its underlying neurobiology and pathophysiology. Obtaining this knowledge largely relies on animal studies. Thus, choosing the appropriate animal model is one of the most critical steps in pre-clinical medication development. Among the range of animal models that have been used to investigate excessive alcohol consumption in rodents, the postdependent model stands out. It was specifically developed to test the role of negative affect as a key driving force in a perpetuating addiction cycle for alcoholism. Here, we will describe our approach to make rats dependent via chronic intermittent exposure to alcohol, discuss the validity of this model, and compare it with other commonly used animal models of alcoholism. We will summarize evidence that postdependent rats fulfill several criteria of a 'Diagnostic and Statistical Manual of Mental Disorders IV/V-like' diagnostic system. Importantly, these animals show long-lasting excessive consumption of and increased motivation for alcohol, and evidence for loss of control over alcohol intake. Our conclusion that postdependent rats are an excellent model for medication development for alcoholism is underscored by a summary of more than two dozen pharmacological tests aimed at reversing these abnormal alcohol responses. We will end with open questions on the use of this model. In the tradition of the Sanchis-Segura and Spanagel review, we provide comic strips that illustrate the postdependent procedure and relevant phenotypes in this review.
Collapse
Affiliation(s)
| | - Wolfgang H. Sommer
- Institute of Psychopharmacology; University of Heidelberg; Germany
- Department of Addiction Medicine; Central Institute of Mental Health; Medical Faculty Mannheim; University of Heidelberg; Germany
| |
Collapse
|
11
|
Grisel JE, Beasley JB, Bertram EC, Decker BE, Duan CA, Etuma M, Hand A, Locklear MN, Whitmire MP. Initial subjective reward: single-exposure conditioned place preference to alcohol in mice. Front Neurosci 2014; 8:345. [PMID: 25408633 PMCID: PMC4219544 DOI: 10.3389/fnins.2014.00345] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 10/09/2014] [Indexed: 11/21/2022] Open
Abstract
Most adults consume alcohol with relative impunity, but about 10–20% of users persist (or progress) in their consumption, despite mounting and serious repercussions. Identifying at-risk individuals before neuroadaptative changes associated with chronic use become well ingrained is thus a key step in mitigating and preventing the end stage disease and its devastating impacts. Explaining liability has been impeded, in part, by the absence of animal models for assessing initial sensitivity to the drug's reinforcing properties, an important endophenotype in the trajectory toward excessive drinking. Here we assess the initial rewarding effects of the drug in a novel application of the conditioned place preference paradigm. In contrast to previous studies that have all employed repeated drug administration, we demonstrated a robust preference for a context paired with a single exposure to 1.5 g/kg EtOH in male and female subjects of three strains. This model validates an assay of initial sensitivity to the subjective rewarding effects of alcohol, a widely used drug with multifarious impacts on both brain and society, and provides a new tool for theory-driven endophenotypic pharmacogenetic approaches to understanding and treating addiction.
Collapse
Affiliation(s)
| | | | | | | | - Chunyu A Duan
- Neuroscience, Furman University Greenville, SC, USA ; Princeton Neuroscience Institute, Princeton University Princeton, NJ, USA
| | - Mahder Etuma
- Neuroscience, Bucknell University Lewisburg, PA, USA
| | - Annie Hand
- Neuroscience, Furman University Greenville, SC, USA
| | - Mallory N Locklear
- Neuroscience, Furman University Greenville, SC, USA ; Neurology and Behavior, Stony Brook University, State University of New York Stony Brook, NY, USA
| | | |
Collapse
|
12
|
Ghazal P, Corsi M, Roth A, Faggioni F, Corti C, Merlo Pick E, Pucciarelli S, Ciccocioppo R, Ubaldi M. Paradoxical response to the sedative effects of diazepam and alcohol in C57BL/6J mice lacking the neuropeptide S receptor. Peptides 2014; 61:107-13. [PMID: 25240770 DOI: 10.1016/j.peptides.2014.09.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/09/2014] [Accepted: 09/09/2014] [Indexed: 10/24/2022]
Abstract
The neuropeptide S (NPS) system is characterized by a unique pharmacology because it has anxiolytic-like effects and promotes arousal and wakefulness. To shed light on this peptidergic system, we tested the sedative effect of the central depressants diazepam and ethanol on the loss of righting reflex in mice lacking the neuropeptide S receptor (NPSR), NPSR(-/-). Furthermore, we tested the effect of the intracerebroventricular (ICV) administration of NPS on the sedative effect of diazepam and ethanol in NPSR(-/-) and their wild type counterpart NPSR(+/+). Finally, we evaluated the effect of the pro-arousal neuropeptides CRF and Hcrt-1/Ox-A in NPSR-deficient mice. Contrary to our expectations, the results showed that the NPSR(-/-) were less sensitive to the hypnotic effects of both diazepam and ethanol compared with their wild type littermates. ICV NPS was able to attenuate the sedative effect of both alcohol and diazepam in wild type mice, but not in the NPSR(-/-) line. The administration of CRF and Hcrt-1/Ox-A, two classic pro-arousal peptides, elicited the same effects in both NPSR(-/-) and wild type mice, ruling out the possibility that adaptive mechanisms occurring at the level of these two systems could have occurred during NPSR(-/-) development to compensate for the lack of NPSR receptors. Our findings demonstrated that the deletion of NPSR leads to minor changes in the arousal behavior of mice. Moreover, we demonstrated that the deletion of NPSR did not lead to compensatory changes in the vigilance-promoting effects of the CRF and Hcrt-1/Ox-A systems.
Collapse
Affiliation(s)
- Pasha Ghazal
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, MC, Italy
| | - Mauro Corsi
- Neurosciences Centre of Excellence for Drug Discovery, GlaxoSmithKline, Medicines Research Centre, 37135 Verona, Italy
| | - Adelheid Roth
- Neurosciences Centre of Excellence for Drug Discovery, GlaxoSmithKline, Medicines Research Centre, 37135 Verona, Italy
| | - Federico Faggioni
- Neurosciences Centre of Excellence for Drug Discovery, GlaxoSmithKline, Medicines Research Centre, 37135 Verona, Italy
| | - Corrado Corti
- Neurosciences Centre of Excellence for Drug Discovery, GlaxoSmithKline, Medicines Research Centre, 37135 Verona, Italy
| | - Emilio Merlo Pick
- Neurosciences Centre of Excellence for Drug Discovery, GlaxoSmithKline, Medicines Research Centre, 37135 Verona, Italy
| | - Sandra Pucciarelli
- School of Biosciences and Biotechnology, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, MC, Italy
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, MC, Italy.
| |
Collapse
|
13
|
Ciccocioppo R, Stopponi S, Economidou D, Kuriyama M, Kinoshita H, Heilig M, Roberto M, Weiss F, Teshima K. Chronic treatment with novel brain-penetrating selective NOP receptor agonist MT-7716 reduces alcohol drinking and seeking in the rat. Neuropsychopharmacology 2014; 39:2601-10. [PMID: 24863033 PMCID: PMC4207340 DOI: 10.1038/npp.2014.113] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 03/31/2014] [Accepted: 04/22/2014] [Indexed: 11/09/2022]
Abstract
Since its discovery, the nociceptin/orphanin FQ (N/OFQ)-NOP receptor system has been extensively investigated as a promising target to treat alcoholism. Encouraging results obtained with the endogenous ligand N/OFQ stimulated research towards the development of novel brain-penetrating NOP receptor agonists with a pharmacological and toxicological profile compatible with clinical development. Here we describe the biochemical and alcohol-related behavioral effects of the novel NOP receptor agonist MT-7716. MT-7716 has high affinity for human NOP receptors expressed in HEK293 cells with a Ki value of 0.21 nM. MT-7716 concentration-dependently stimulated GTPγ(35)S binding with an EC50 value of 0.30 nM and its efficacy was similar to N/OFQ, suggesting that MT7716 is a full agonist at NOP receptors. In the two bottle choice test MT-7716 (0, 0.3, 1, and 3 mg/kg, bid) given orally for 14 days dose-dependently decreased voluntary alcohol intake in Marchigian Sardinian rats. The effect became gradually stronger following repeated administration, and was still significant 1 week after discontinuation of the drug. Oral naltrexone (30 mg/kg, bid) for 14 days also reduced ethanol intake; however, the effect decreased over the treatment period and rapidly disappeared when drug treatment was discontinued. MT-7716 is also effective for preventing reinstatement caused by both ethanol-associated environmental stimuli and stress. Finally, to investigate the effect of MT-7716 on alcohol withdrawal symptoms, Wistar rats were withdrawn from a 7-day alcohol liquid diet. MT-7716 significantly attenuated somatic alcohol withdrawal symptoms. Together these findings indicate that MT-7716 is a promising candidate for alcoholism treatment remaining effective with chronic administration.
Collapse
Affiliation(s)
- Roberto Ciccocioppo
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Serena Stopponi
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Daina Economidou
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Makoto Kuriyama
- Department II (CNS), Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Hiroshi Kinoshita
- Department II (CNS), Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Markus Heilig
- Laboratory of Clinical and Translational Studies, National Institute of Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Friedbert Weiss
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Koji Teshima
- Department II (CNS), Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| |
Collapse
|
14
|
Ryan PJ, Krstew EV, Sarwar M, Gundlach AL, Lawrence AJ. Relaxin-3 mRNA levels in nucleus incertus correlate with alcohol and sucrose intake in rats. Drug Alcohol Depend 2014; 140:8-16. [PMID: 24837581 DOI: 10.1016/j.drugalcdep.2014.04.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/02/2014] [Accepted: 04/09/2014] [Indexed: 11/17/2022]
Abstract
BACKGROUND Chronic alcohol intake produces multiple neuroadaptive changes, including up- and down-regulation of neuropeptides and receptors. There are widespread projections of relaxin-3 containing neurons to, and abundant relaxin family peptide 3 receptor (RXFP3) expression within, brain regions involved in modulating alcohol intake. Recently we demonstrated the involvement of relaxin-3/RXFP3 signalling in alcohol-seeking in rats; therefore in this study we examined whether relaxin-3 and/or RXFP3 expression were altered by chronic alcohol intake in alcohol-preferring iP rats. METHODS Expression of relaxin-3 mRNA in the hindbrain nucleus incertus and RXFP3 radioligand binding levels in discrete forebrain regions were investigated following voluntary intake of alcohol or sucrose for 12 weeks, with a 2 day washout, using quantitative in situ hybridisation histochemistry and in vitro receptor autoradiography, respectively, in cohorts of adult, male iP rats. RESULTS Levels of relaxin-3 mRNA in the hindbrain nucleus incertus were positively correlated with the level of intake of both alcohol (r(12)=0.59, p=0.03) and sucrose (r(7)=0.70, p=0.04) in iP rats. Dense binding of the RXFP3-selective radioligand, [(125)]-R3/I5, was detected in hypothalamic and extrahypothalamic sites, but no significant changes in the density of RXFP3 were observed in the brain regions quantified following chronic sucrose or ethanol intake. CONCLUSIONS Our findings suggest high endogenous relaxin-3 expression may be associated with higher intake of rewarding substances, rather than its expression being regulated in response to their intake, consistent with an active role for the relaxin-3/RXFP3 system in modulating ingestive and alcohol-related behaviours.
Collapse
Affiliation(s)
- P J Ryan
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - E V Krstew
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - M Sarwar
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Parkville, Victoria, Australia; Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - A L Gundlach
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia; Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Victoria, Australia
| | - A J Lawrence
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
15
|
Kallupi M, Varodayan FP, Oleata CS, Correia D, Luu G, Roberto M. Nociceptin/orphanin FQ decreases glutamate transmission and blocks ethanol-induced effects in the central amygdala of naive and ethanol-dependent rats. Neuropsychopharmacology 2014; 39:1081-92. [PMID: 24169802 PMCID: PMC3957102 DOI: 10.1038/npp.2013.308] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 10/08/2013] [Accepted: 10/24/2013] [Indexed: 12/11/2022]
Abstract
The central nucleus of the amygdala (CeA) mediates several addiction-related processes and nociceptin/orphanin FQ (nociceptin) regulates ethanol intake and anxiety-like behaviors. Glutamatergic synapses, in the CeA and throughout the brain, are very sensitive to ethanol and contribute to alcohol reinforcement, tolerance, and dependence. Previously, we reported that in the rat CeA, acute and chronic ethanol exposures significantly decrease glutamate transmission by both pre- and postsynaptic actions. In this study, using electrophysiological techniques in an in vitro CeA slice preparation, we investigated the effects of nociceptin on glutamatergic transmission and its interaction with acute ethanol in naive and ethanol-dependent rats. We found that nociceptin (100-1000 nM) diminished basal-evoked compound glutamatergic receptor-mediated excitatory postsynaptic potentials (EPSPs) and spontaneous and miniature EPSCs (s/mEPSCs) by mainly decreasing glutamate release in the CeA of naive rats. Notably, nociceptin blocked the inhibition induced by acute ethanol (44 mM) and ethanol blocked the nociceptin-induced inhibition of evoked EPSPs in CeA neurons of naive rats. In neurons from chronic ethanol-treated (ethanol-dependent) rats, the nociceptin-induced inhibition of evoked EPSP amplitude was not significantly different from that in naive rats. Application of [Nphe1]Nociceptin(1-13)NH2, a nociceptin receptor (NOP) antagonist, revealed tonic inhibitory activity of NOP on evoked CeA glutamatergic transmission only in ethanol-dependent rats. The antagonist also blocked nociceptin-induced decreases in glutamatergic responses, but did not affect ethanol-induced decreases in evoked EPSP amplitude. Taken together, these studies implicate a potential role for the nociceptin system in regulating glutamatergic transmission and a complex interaction with ethanol at CeA glutamatergic synapses.
Collapse
Affiliation(s)
- Marsida Kallupi
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Florence P Varodayan
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Christopher S Oleata
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Diego Correia
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
- Department of Pharmacology, Universidade Federal do Paraná, Jardim das Américas, Curitiba, Paraná, Brazil
| | - George Luu
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
16
|
Kallupi M, Oleata CS, Luu G, Teshima K, Ciccocioppo R, Roberto M. MT-7716, a novel selective nonpeptidergic NOP receptor agonist, effectively blocks ethanol-induced increase in GABAergic transmission in the rat central amygdala. Front Integr Neurosci 2014; 8:18. [PMID: 24600360 PMCID: PMC3927450 DOI: 10.3389/fnint.2014.00018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 02/01/2014] [Indexed: 11/13/2022] Open
Abstract
The GABAergic system in the central amygdala (CeA) plays a major role in ethanol dependence and the anxiogenic-like response to ethanol withdrawal. A large body of evidence shows that Nociceptin/Orphanin FQ (N/OFQ) regulates ethanol intake and anxiety-like behavior. In the rat, ethanol significantly augments CeA GABA release, whereas N/OFQ diminishes it. Using electrophysiological techniques in an in vitro slice preparation, in this study we investigated the effects of a nonpeptidergic NOP receptor agonist, MT-7716 [(R)-2-3-[1-(Acenaphthen-1-yl)piperidin-4-yl]-2-oxo-2,3-dihydro-1H-benzimidazol-1-yl-N-methylacetamide hydrochloride hydrate], and its interaction with ethanol on GABAergic transmission in CeA slices of naïve rats. We found that MT-7716 dose-dependently (100-1000 nM) diminished evoked GABAA receptor-mediated inhibitory postsynaptic potentials (IPSPs) and increased paired-pulse facilitation (PPF) ratio of these evoked IPSPs, suggesting a presynaptic site of action of the MT-7716 by decreasing GABA release at CeA synapses. The presynaptic action of MT-7716 was also supported by the significant decrease in the frequency of miniature inhibitory postsynaptic currents (mIPSCs) induced by the nociceptin receptor (NOP) agonist. Interestingly, MT-7716 prevented the ethanol-induced augmentation of evoked IPSPs. A putative selective NOP antagonist, [Nphe1]Nociceptin(1-13)NH2, totally prevented the MT-7716-induced inhibition of IPSP amplitudes indicating that MT-7716 exerts its effect through NOPs. These data provide support for an interaction between the nociceptin and GABAergic systems in the CeA and for the anti-alcohol properties of the NOP activation. The development of a synthetic nonpeptidergic NOP receptor agonist such as MT-7716 may represent a useful therapeutic target for alcoholism.
Collapse
Affiliation(s)
- Marsida Kallupi
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La JollaCA, USA
- Pharmacology Unit, School of Pharmacy, University of CamerinoCamerino, Italy
| | - Christopher S. Oleata
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La JollaCA, USA
| | - George Luu
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La JollaCA, USA
| | - Koji Teshima
- Department II (CNS), Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma CorporationYokohama, Japan
| | - Roberto Ciccocioppo
- Pharmacology Unit, School of Pharmacy, University of CamerinoCamerino, Italy
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La JollaCA, USA
| |
Collapse
|