Endogenous opioid system in addiction and addiction-related behaviors

Current Opinion in Behavioral Sciences - Tập 13 - Trang 196-202 - 2017
Brian Reed1, Eduardo R Butelman1, Mary Jeanne Kreek1
1Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA

Tài liệu tham khảo

Kreek, 1984, ACTH, cortisol and beta-endorphin response to metyrapone testing during chronic methadone maintenance treatment in humans, Neuropeptides, 5, 277, 10.1016/0143-4179(84)90081-7 Goldman, 2005, The genetics of addictions: uncovering the genes, Nat Rev Genet, 6, 521, 10.1038/nrg1635 Reed, 2014, Genetics of opiate addiction, Curr Psychiatry Rep, 16, 504, 10.1007/s11920-014-0504-6 Bond, 1998, Single-nucleotide polymorphism in the human mu opioid receptor gene alters beta-endorphin binding and activity: possible implications for opiate addiction, Proc Natl Acad Sci U S A, 95, 9608, 10.1073/pnas.95.16.9608 Levran, 2012, The genetics of the opioid system and specific drug addictions, Hum Genet, 131, 823, 10.1007/s00439-012-1172-4 Mague, 2009, Mouse model of OPRM1 (A118G) polymorphism has sex-specific effects on drug-mediated behavior, Proc Natl Acad Sci U S A, 106, 10847, 10.1073/pnas.0901800106 Zhang, 2015, Mouse model of the OPRM1 (A118G) polymorphism: differential heroin self-administration behavior compared with wild-type mice, Neuropsychopharmacology, 40, 1091, 10.1038/npp.2014.286 Ramchandani, 2011, A genetic determinant of the striatal dopamine response to alcohol in men, Mol Psychiatry, 16, 809, 10.1038/mp.2010.56 Robinson, 2015, Receptor reserve moderates mesolimbic responses to opioids in a humanized mouse model of the OPRM1 A118G polymorphism, Neuropsychopharmacology, 40, 2614, 10.1038/npp.2015.109 Henderson-Redmond, 2016, Morphine-induced antinociception and reward in “humanized” mice expressing the mu opioid receptor A118G polymorphism, Brain Res Bull, 123, 5, 10.1016/j.brainresbull.2015.10.007 Crowley, 2016, Dynorphin controls the gain of an amygdalar anxiety circuit, Cell Rep, 14, 2774, 10.1016/j.celrep.2016.02.069 Tejeda, 2015, Prefrontal cortical kappa opioid receptors attenuate responses to amygdala inputs, Neuropsychopharmacology, 40, 2856, 10.1038/npp.2015.138 Al-Hasani, 2015, Distinct subpopulations of nucleus accumbens dynorphin neurons drive aversion and reward, Neuron, 87, 1063, 10.1016/j.neuron.2015.08.019 Barish, 2013, Design and functional evaluation of an optically active μ-opioid receptor, Eur J Pharmacol, 705, 42, 10.1016/j.ejphar.2013.01.065 Siuda, 2015, Spatiotemporal control of opioid signaling and behavior, Neuron, 86, 923, 10.1016/j.neuron.2015.03.066 Coward, 1998, Controlling signaling with a specifically designed Gi-coupled receptor, Proc Natl Acad Sci U S A, 95, 352, 10.1073/pnas.95.1.352 Koch, 2015, Hypothalamic POMC neurons promote cannabinoid-induced feeding, Nature, 519, 45, 10.1038/nature14260 Vardy, 2015, A new DREADD facilitates the multiplexed chemogenetic interrogation of behavior, Neuron, 86, 936, 10.1016/j.neuron.2015.03.065 Rorick-Kehn, 2014, LY2456302 is a novel, potent, orally-bioavailable small molecule kappa-selective antagonist with activity in animal models predictive of efficacy in mood and addictive disorders, Neuropharmacology, 77, 131, 10.1016/j.neuropharm.2013.09.021 Anderson, 2016, Stress-induced enhancement of ethanol intake in C57BL/6J mice with a history of chronic ethanol exposure: involvement of kappa opioid receptors, Front Cell Neurosci, 10, 45, 10.3389/fncel.2016.00045 Grimwood, 2011, Pharmacological characterization of 2-methyl-N-((2′-(pyrrolidin-1-ylsulfonyl)biphenyl-4-yl)methyl)propan-1-amine (PF-04455242), a high-affinity antagonist selective for κ-opioid receptors, J Pharmacol Exp Ther, 339, 555, 10.1124/jpet.111.185108 Aldrich, 2009, Zyklophin, a systemically active selective kappa opioid receptor peptide antagonist with short duration of action, Proc Natl Acad Sci U S A, 106, 18396, 10.1073/pnas.0910180106 Reed, 2012, Chromatin alterations in response to forced swimming underlie increased prodynorphin transcription, Neuroscience, 220, 109, 10.1016/j.neuroscience.2012.06.006 Spangler, 1993, ‘Binge’ cocaine administration induces a sustained increase of prodynorphin mRNA in rat caudate-putamen, Brain Res Mol Brain Res, 19, 323, 10.1016/0169-328X(93)90133-A Daunais, 1995, Cocaine binges differentially alter striatal preprodynorphin and zif/268 mRNAs, Brain Res Mol Brain Res, 29, 201, 10.1016/0169-328X(94)00246-B Lowe, 2014, Safety, tolerability, and pharmacokinetic evaluation of single- and multiple-ascending doses of a novel kappa opioid receptor antagonist LY2456302 and drug interaction with ethanol in healthy subjects, J Clin Pharmacol, 54, 968, 10.1002/jcph.286 Korpi, 2016, Continuous delivery of naltrexone and nalmefene leads to tolerance in reducing alcohol drinking and to supersensitivity of brain opioid receptors, Addict Biol Soyka, 2016, Comparing Nalmefene and Naltrexone in alcohol dependence: are there any differences? Results from an indirect meta analysis, Pharmacopsychiatry, 49, 66, 10.1055/s-0035-1565184 Galloway, 2005, Pharmacokinetics, safety, and tolerability of a depot formulation of naltrexone in alcoholics: an open-label trial, BMC Psychiatry, 5, 18, 10.1186/1471-244X-5-18 Feeney, 2001, Adherence with naltrexone prescription advice in hospital outpatient alcohol rehabilitation programme, J Clin Pharm Ther, 26, 73, 10.1046/j.1365-2710.2001.00326.x Besirli, 2014, Hypothalamic–pituitary–adrenal axis response to oral naltrexone in alcoholics during early withdrawal, Pharmacopsychiatry, 47, 151, 10.1055/s-0034-1381983 Schluger, 1998, Nalmefene causes greater hypothalamic–pituitary–adrenal axis activation than naloxone in normal volunteers: implications for the treatment of alcoholism, Alcohol Clin Exp Res, 22, 1430, 10.1111/j.1530-0277.1998.tb03931.x Butelman, 2012, κ-opioid receptor/dynorphin system: genetic and pharmacotherapeutic implications for addiction, Trends Neurosci, 35, 587, 10.1016/j.tins.2012.05.005 Raehal, 2005, Morphine side effects in beta-arrestin 2 knockout mice, J Pharmacol Exp Ther, 314, 1195, 10.1124/jpet.105.087254 Violin, 2014, Biased ligands at G-protein-coupled receptors: promise and progress, Trends Pharmacol Sci, 35, 308, 10.1016/j.tips.2014.04.007 Kenakin, 2015, The effective application of biased signaling to new drug discovery, Mol Pharmacol, 88, 1055, 10.1124/mol.115.099770 Winpenny, 2016, Biased ligand quantification in drug discovery: from theory to high throughput screening to identify new biased μ opioid receptor agonists, Br J Pharmacol, 173, 1393, 10.1111/bph.13441 Yang, 2014, Morphine and DAMGO produce an opposite effect on presynaptic glutamate release via different downstream pathways of μ opioid receptors in the basolateral amygdala, Neuropharmacology, 86, 353, 10.1016/j.neuropharm.2014.08.021 Chen, 2013, Structure–activity relationships and discovery of a G protein biased μ opioid receptor ligand, [(3-methoxythiophen-2-yl)methyl]({2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro [4.5]decan-9-yl]ethyl})amine (TRV130), for the treatment of acute severe pain, J Med Chem, 56, 8019, 10.1021/jm4010829 DeWire, 2013, A G protein biased ligand at the μ-opioid receptor is potently analgesic with reduced gastrointestinal and respiratory dysfunction compared with morphine, J Pharmacol Exp Ther, 344, 708, 10.1124/jpet.112.201616 Soergel, 2014, Biased agonism of the μ-opioid receptor by TRV130 increases analgesia and reduces on-target adverse effects versus morphine: a randomized, double-blind, placebo-controlled, crossover study in healthy volunteers, Pain, 155, 1829, 10.1016/j.pain.2014.06.011 Thompson, 2015, Biased agonism of endogenous opioid peptides at the μ-opioid receptor, Mol Pharmacol, 88, 335, 10.1124/mol.115.098848 Schmid, 2013, Functional selectivity of 6′-guanidinonaltrindole (6′-GNTI) at κ-opioid receptors in striatal neurons, J Biol Chem, 288, 22387, 10.1074/jbc.M113.476234 Dogra, 2015, Biased agonism at kappa opioid receptors: implication in pain and mood disorders, Eur J Pharmacol, 763, 184, 10.1016/j.ejphar.2015.07.018 Charfi, 2014, Ligand- and cell-dependent determinants of internalization and cAMP modulation by delta opioid receptor (DOR) agonists, Cell Mol Life Sci, 71, 1529, 10.1007/s00018-013-1461-7 Pradhan, 2010, Ligand-directed trafficking of the δ-opioid receptor in vivo: two paths toward analgesic tolerance, J Neurosci, 30, 16459, 10.1523/JNEUROSCI.3748-10.2010 Pradhan, 2016, Agonist-specific recruitment of arrestin isoforms differentially modify delta opioid receptor function, J Neurosci, 36, 3541, 10.1523/JNEUROSCI.4124-15.2016 White, 2014, Identification of novel functionally selective κ-opioid receptor scaffolds, Mol Pharmacol, 85, 83, 10.1124/mol.113.089649 Zhou, 2013, Development of functionally selective, small molecule agonists at kappa opioid receptors, J Biol Chem, 288, 36703, 10.1074/jbc.M113.504381 White, 2015, The G protein-biased κ-opioid receptor agonist RB-64 is analgesic with a unique spectrum of activities in vivo, J Pharmacol Exp Ther, 352, 98, 10.1124/jpet.114.216820 Stahl, 2015, A novel method for analyzing extremely biased agonism at G protein-coupled receptors, Mol Pharmacol, 87, 866, 10.1124/mol.114.096503 Lovell, 2015, Structure–activity relationship studies of functionally selective kappa opioid receptor agonists that modulate ERK 1/2 phosphorylation while preserving G protein over βarrestin2 signaling bias, ACS Chem Neurosci, 6, 1411, 10.1021/acschemneuro.5b00092 Granier, 2012, Structure of the δ-opioid receptor bound to naltrindole, Nature, 485, 400, 10.1038/nature11111 Huang, 2015, Structural insights into μ-opioid receptor activation, Nature, 524, 315, 10.1038/nature14886 Manglik, 2012, Crystal structure of the μ-opioid receptor bound to a morphinan antagonist, Nature, 485, 321, 10.1038/nature10954 Wu, 2012, Structure of the human κ-opioid receptor in complex with JDTic, Nature, 485, 327, 10.1038/nature10939 Goldfeld, 2015, Docking and free energy perturbation studies of ligand binding in the kappa opioid receptor, J Phys Chem B, 119, 824, 10.1021/jp5053612 Martinez-Mayorga, 2013, Ligand/kappa-opioid receptor interactions: insights from the X-ray crystal structure, Eur J Med Chem, 66, 114, 10.1016/j.ejmech.2013.05.021 Negri, 2013, Discovery of a novel selective kappa-opioid receptor agonist using crystal structure-based virtual screening, J Chem Inf Model, 53, 521, 10.1021/ci400019t Vardy, 2013, Chemotype-selective modes of action of κ-opioid receptor agonists, J Biol Chem, 288, 34470, 10.1074/jbc.M113.515668