249
Views
2
CrossRef citations to date
0
Altmetric
Review

Influence of the OPRM1 A118G Polymorphism on Alcohol-Induced Euphoria, Risk for Alcoholism and the Clinical Efficacy of Naltrexone

, , &
Pages 1161-1172 | Published online: 21 Aug 2012

References

  • Rehm J , MathersC, PopovaS, ThavorncharoensapM, TeerawattananonY, PatraJ. Global burden of disease and injury and economic cost attributable to alcohol use and alcohol-use disorders. Lancet373(9682) , 2223–2233 (2009).
  • Heilig M , GoldmanD, BerrettiniW, O‘BrienCP. Pharmacogenetic approaches to the treatment of alcohol addiction. Nat. Rev. Neurosci.12(11) , 670–684 (2011).
  • Pert CB , SnyderSH. Opiate receptor: demonstration in nervous tissue. Science179(4077) , 1011–1014 (1973).
  • Akil H , MayerDJ, LiebeskindJC. Antagonism of stimulation-produced analgesia by naloxone, a narcotic antagonist. Science191(4230) , 961–962 (1976).
  • Hughes J , SmithTW, KosterlitzHW, FothergillLA, MorganBA, MorrisHR. Identification of two related pentapeptides from the brain with potent opiate agonist activity. Nature258(5536) , 577–580 (1975).
  • Goldstein A , TachibanaS, LowneyLI, HunkapillerM, HoodL. Dynorphin-(1–13), an extraordinarily potent opioid peptide. Proc. Natl Acad. Sci. USA76(12) , 6666–6670 (1979).
  • Le Merrer J , BeckerJA, BefortK, KiefferBL. Reward processing by the opioid system in the brain. Physiol. Rev.89(4) , 1379–1412 (2009).
  • Mague SD , BlendyJA. OPRM1 SNP (A118G): involvement in disease development, treatment response, and animal models. Drug Alcohol Depend.108(3) , 172–182 (2010).
  • Shabalina SA , ZaykinDV, GrisP et al. Expansion of the human mu-opioid receptor gene architecture: novel functional variants. Hum. Mol. Genet. 18(6) , 1037–1051 (2009).
  • Bergen AW , KokoszkaJ, PetersonR et al. Mu opioid receptor gene variants: lack of association with alcohol dependence. Mol. Psychiatry 2(6) , 490–494 (1997).
  • Gelernter J , KranzlerH, CubellsJ. Genetics of two mu opioid receptor gene (OPRM1) exon I polymorphisms: population studies, and allele frequencies in alcohol- and drug-dependent subjects. Mol. Psychiatry4(5) , 476–483 (1999).
  • Ray LA , BujarskiS, ChinPF, MiottoK. Pharmacogenetics of naltrexone in Asian Americans: a randomized placebo-controlled laboratory study. Neuropsychopharmacology37(2) , 445–455 (2012).
  • Bond C , LaforgeKS, TianM et al. 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. USA 95(16) , 9608–9613 (1998).
  • Miller GM , BendorJ, TiefenbacherS, YangH, NovakMA, MadrasBK. A mu-opioid receptor single nucleotide polymorphism in rhesus monkey: association with stress response and aggression. Mol. Psychiatry9(1) , 99–108 (2004).
  • Vallender EJ , PriddyCM, ChenGL, MillerGM. Human expression variation in the mu-opioid receptor is paralleled in rhesus macaque. Behav. Genet.38(4) , 390–395 (2008).
  • Beyer A , KochT, SchroderH, SchulzS, HolltV. Effect of the A118G polymorphism on binding affinity, potency and agonist-mediated endocytosis, desensitization, and resensitization of the human mu-opioid receptor. J. Neurochem.89(3) , 553–560 (2004).
  • Zhang Y , WangD, JohnsonAD, PappAC, SadeeW. Allelic expression imbalance of human mu opioid receptor (OPRM1) caused by variant A118G. J. Biol. Chem.280(38) , 32618–32624 (2005).
  • Kroslak T , LaforgeKS, GianottiRJ, HoA, NielsenDA, KreekMJ. The single nucleotide polymorphism A118G alters functional properties of the human mu opioid receptor. J. Neurochem.103(1) , 77–87 (2007).
  • Mague SD , IsiegasC, HuangP, Liu-ChenLY, LermanC, BlendyJA. Mouse model of OPRM1 (A118G) polymorphism has sex-specific effects on drug-mediated behavior. Proc. Natl Acad. Sci. USA106(26) , 10847–10852 (2009).
  • Wang YJ , HuangP, UngA, BlendyJA, Liu-ChenLY. Reduced expression of the mu opioid receptor in some, but not all, brain regions in mice with Oprm1 A112G. Neuroscience205 , 178–184 (2012).
  • Huang P , ChenC, MagueSD, BlendyJA, Liu-ChenLY. A common single nucleotide polymorphism A118G of the mu opioid receptor alters its N-glycosylation and protein stability. Biochem. J.441(1) , 379–386 (2012).
  • Weerts EM , MccaulME, KuwabaraH et al. Influence of OPRM1 Asn40Asp variant (A118G) on [11C]carfentanil binding potential: preliminary findings in human subjects. Int. J. Neuropsychopharmacol. 8 , 1–7 (2012).
  • Marinelli PW , BaiL, QuirionR, GianoulakisC. A microdialysis profile of Met-enkephalin release in the rat nucleus accumbens following alcohol administration. Alcohol Clin. Exp. Res.29(10) , 1821–1828 (2005).
  • Marinelli PW , QuirionR, GianoulakisC. A microdialysis profile of beta-endorphin and catecholamines in the rat nucleus accumbens following alcohol administration. Psychopharmacology (Berl.)169(1) , 60–67 (2003).
  • Lam MP , GianoulakisC. Effects of acute ethanol on corticotropin-releasing hormone and beta-endorphin systems at the level of the rat central amygdala. Psychopharmacology (Berl.)218(1) , 229–239 (2011).
  • Jarjour S , BaiL, GianoulakisC. Effect of acute ethanol administration on the release of opioid peptides from the midbrain including the ventral tegmental area. Alcohol Clin. Exp. Res.33(6) , 1033–1043 (2009).
  • Volpicelli JR , UlmRR, HopsonN. Alcohol drinking in rats during and following morphine injections. Alcohol8(4) , 289–292 (1991).
  • Zhang M , KelleyAE. Intake of saccharin, salt, and ethanol solutions is increased by infusion of a mu opioid agonist into the nucleus accumbens. Psychopharmacology (Berl.)159(4) , 415–423 (2002).
  • Hall FS , SoraI, UhlGR. Ethanol consumption and reward are decreased in mu-opiate receptor knockout mice. Psychopharmacology (Berl.)154(1) , 43–49 (2001).
  • Roberts AJ , McdonaldJS, HeyserCJ et al. mu-opioid receptor knockout mice do not self-administer alcohol. J. Pharmacol. Exp. Ther. 293(3) , 1002–1008 (2000).
  • Gianoulakis C , KrishnanB, ThavundayilJ. Enhanced sensitivity of pituitary beta-endorphin to ethanol in subjects at high risk of alcoholism. Arch. Gen. Psychiatry53(3) , 250–257 (1996).
  • Mitchell JM , O‘NeilJP, JanabiM, MarksSM, JagustWJ, FieldsHL. Alcohol consumption induces endogenous opioid release in the human orbitofrontal cortex and nucleus accumbens. Sci. Transl. Med.4(116) , 116ra6 (2012).
  • Gonzales RA , JobMO, DoyonWM. The role of mesolimbic dopamine in the development and maintenance of ethanol reinforcement. Pharmacol. Ther.103(2) , 121–146 (2004).
  • Herz A . Endogenous opioid systems and alcohol addiction. Psychopharmacology (Berl.)129(2) , 99–111 (1997).
  • Matsui A , WilliamsJT. Opioid-sensitive GABA inputs from rostromedial tegmental nucleus synapse onto midbrain dopamine neurons. J. Neurosci.31(48) , 17729–17735 (2011).
  • Fallon JH , LeslieFM, ConeRI. Dynorphin-containing pathways in the substantia nigra and ventral tegmentum: a double labeling study using combined immunofluorescence and retrograde tracing. Neuropeptides5(4–6) , 457–460 (1985).
  • Leyton M , RajabiH, StewartJ. U-50,488H into A10 reduces haloperidol-induced elevations of accumbens dopamine. Neuroreport3(12) , 1127–1130 (1992).
  • Devine DP , LeoneP, PocockD, WiseRA. Differential involvement of ventral tegmental mu, delta and kappa opioid receptors in modulation of basal mesolimbic dopamine release: in vivo microdialysis studies. J. Pharmacol. Exp. Ther.266(3) , 1236–1246 (1993).
  • Margolis EB , HjelmstadGO, BonciA, FieldsHL. Kappa-opioid agonists directly inhibit midbrain dopaminergic neurons. J. Neurosci.23(31) , 9981–9986 (2003).
  • Roth-Deri I , ZangenA, AleliM et al. Effect of experimenter-delivered and self-administered cocaine on extracellular beta-endorphin levels in the nucleus accumbens. J. Neurochem. 84(5) , 930–938 (2003).
  • Kalivas PW , WiderlovE, StanleyD, BreeseG, PrangeAJ Jr. Enkephalin action on the mesolimbic system: a dopamine-dependent and a dopamine-independent increase in locomotor activity. J. Pharmacol. Exp. Ther.227(1) , 229–237 (1983).
  • Ray LA , HutchisonKE. A polymorphism of the mu-opioid receptor gene (OPRM1) and sensitivity to the effects of alcohol in humans. Alcohol Clin. Exp. Res.28(12) , 1789–1795 (2004).
  • Ray LA , HutchisonKE. Effects of naltrexone on alcohol sensitivity and genetic moderators of medication response: a double-blind placebo-controlled study. Arch. Gen. Psychiatry64(9) , 1069–1077 (2007).
  • Ray LA , MirandaR Jr, Tidey JW et al. Polymorphisms of the mu-opioid receptor and dopamine D4 receptor genes and subjective responses to alcohol in the natural environment. J. Abnorm. Psychol.119(1) , 115–125 (2010).
  • Wiers RW , RinckM, DictusM, Van Den Wildenberg E. Relatively strong automatic appetitive action-tendencies in male carriers of the OPRM1 G-allele. Genes Brain Behav.8(1) , 101–106 (2009).
  • Van Den Wildenberg E , WiersRW, DessersJ et al. A functional polymorphism of the mu-opioid receptor gene (OPRM1) influences cue-induced craving for alcohol in male heavy drinkers. Alcohol Clin. Exp. Res. 31(1) , 1–10 (2007).
  • Barr CS , SchwandtM, LindellSG et al. Association of a functional polymorphism in the mu-opioid receptor gene with alcohol response and consumption in male rhesus macaques. Arch. Gen. Psychiatry 64(3) , 369–376 (2007).
  • Vallender EJ , Ruedi-BettschenD, MillerGM, PlattDM. A pharmacogenetic model of naltrexone-induced attenuation of alcohol consumption in rhesus monkeys. Drug Alcohol Depend.109(1–3) , 252–256 (2010).
  • Ramchandani VA , UmhauJ, PavonFJ et al. A genetic determinant of the striatal dopamine response to alcohol in men. Mol. Psychiatry 16(8) , 809–817 (2010).
  • Berridge KC . The debate over dopamine‘s role in reward: the case for incentive salience. Psychopharmacology (Berl.)191(3) , 391–431 (2007).
  • Berridge KC , RobinsonTE, AldridgeJW. Dissecting components of reward: ‘liking‘, ‘wanting‘, and learning. Curr. Opin. Pharmacol.9(1) , 65–73 (2009).
  • McGue M . The behavioural genetics of alcoholism. Curr. Direct Psychol. Sci.8(4) , (1999).
  • McCaul ME , TurkkanJS, SvikisDS, BigelowGE. Familial density of alcoholism: effects on psychophysiological responses to ethanol. Alcohol8(3) , 219–222 (1991).
  • Nagoshi CT , WilsonJR. Influence of family alcoholism history on alcohol metabolism, sensitivity, and tolerance. Alcohol Clin. Exp. Res.11(4) , 392–398 (1987).
  • Schuckit MA . Self-rating of alcohol intoxication by young men with and without family histories of alcoholism. J. Stud. Alcohol41(3) , 242–249 (1980).
  • King AC , De Wit H, Mcnamara PJ, Cao D. Rewarding, stimulant, and sedative alcohol responses and relationship to future binge drinking. Arch. Gen. Psychiatry68(4) , 389–399 (2011).
  • De Waele JP , PapachristouDN, GianoulakisC. The alcohol-preferring C57BL/6 mice present an enhanced sensitivity of the hypothalamic beta-endorphin system to ethanol than the alcohol-avoiding DBA/2 mice. J. Pharmacol. Exp. Ther.261(2) , 788–794 (1992).
  • Li XW , LiTK, FroehlichJC. Enhanced sensitivity of the nucleus accumbens proenkephalin system to alcohol in rats selectively bred for alcohol preference. Brain Res.794(1) , 35–47 (1998).
  • McBride WJ , ChernetE, McKinzieDL, LumengL, Li T-K. Quantitative autoradiography of mu-opioid receptors in the CNS of alcohol-preferring P and -nonpreferring NP rats. Alcohol16(4) , 317–323 (1998).
  • Marinelli PW , KiianmaaK, GianoulakisC. Opioid propeptide mRNA content and receptor density in the brains of AA and ANA rats. Life Sci.66(20) , 1915–1927 (2000).
  • Fadda P , TronciS, ColomboG, FrattaW. Differences in the opioid system in selected brain regions of alcohol-preferring and alcohol-nonpreferring rats. Alcohol Clin. Exp. Res.23(8) , 1296–1305 (1999).
  • Heinz A , ReimoldM, WraseJ et al. Correlation of stable elevations in striatal mu-opioid receptor availability in detoxified alcoholic patients with alcohol craving: a positron emission tomography study using carbon 11-labeled carfentanil. Arch. Gen. Psychiatry 62(1) , 57–64 (2005).
  • Weerts EM , WandGS, KuwabaraH et al. Positron emission tomography imaging of mu- and delta-opioid receptor binding in alcohol-dependent and healthy control subjects. Alcohol Clin. Exp. Res. 35(12) , 2162–2173 (2011).
  • Williams TM , DaviesSJ, TaylorLG et al. Brain opioid receptor binding in early abstinence from alcohol dependence and relationship to craving: an [11C]diprenorphine PET study. Eur. Neuropsychopharmacol. 19(10) , 740–748 (2009).
  • Bart G , KreekMJ, OttJ et al. Increased attributable risk related to a functional mu-opioid receptor gene polymorphism in association with alcohol dependence in central Sweden. Neuropsychopharmacology 30(2) , 417–422 (2005).
  • Miranda R , RayL, JustusA et al. Initial evidence of an association between OPRM1 and adolescent alcohol misuse. Alcohol Clin. Exp. Res. 34(1) , 112–122 (2010).
  • Chen D , LiuL, XiaoY, PengY, YangC, WangZ. Ethnic-specific meta-analyses of association between the OPRM1 A118G polymorphism and alcohol dependence among Asians and Caucasians. Drug Alcohol Depend.123(1–3) , 1–6 (2012).
  • Kim SG . Gender differences in the genetic risk for alcohol dependence – the results of a pharmacogenetic study in Korean alcoholics. Nihon Arukoru Yakubutsu Igakkai Zasshi44(6) , 680–685 (2009).
  • Koller G , ZillP, RujescuD et al. Possible association between OPRM1 genetic variance at the 118 locus and alcohol dependence in a large treatment sample: relationship to alcohol dependence symptoms. Alcohol Clin. Exp. Res. doi:10.1111/j.1530-0277.2011.01714.x (2012) (Epub ahead of print).
  • Nishizawa D , HanW, HasegawaJ et al. Association of mu-opioid receptor gene polymorphism A118G with alcohol dependence in a Japanese population. Neuropsychobiology 53(3) , 137–141 (2006).
  • Kumar D , ChakrabortyJ, DasS. Epistatic effects between variants of kappa-opioid receptor gene and A118G of mu-opioid receptor gene increase susceptibility to addiction in Indian population. Prog. Neuropsychopharmacol. Biol. Psychiatry36(2) , 225–230 (2012).
  • Arias A , FeinnR, KranzlerHR. Association of an Asn40Asp (A118G) polymorphism in the mu-opioid receptor gene with substance dependence: a meta-analysis. Drug Alcohol Depend.83(3) , 262–268 (2006).
  • Sander T , GscheidelN, WendelB et al. Human mu-opioid receptor variation and alcohol dependence. Alcohol Clin. Exp. Res. 22(9) , 2108–2110 (1998).
  • Cloninger CR , BohmanM, SigvardssonS. Inheritance of alcohol abuse. Cross-fostering analysis of adopted men. Arch. Gen. Psychiatry38(8) , 861–868 (1981).
  • Schwandt ML , LindellSG, HigleyJD, SuomiSJ, HeiligM, BarrCS. OPRM1 gene variation influences hypothalamic–pituitary–adrenal axis function in response to a variety of stressors in rhesus macaques. Psychoneuroendocrinology36(9) , 1303–1311 (2011).
  • Chong RY , OswaldL, YangX, UhartM, LinPI, WandGS. The mu-opioid receptor polymorphism A118G predicts cortisol responses to naloxone and stress. Neuropsychopharmacology31(1) , 204–211 (2006).
  • Pratt WM , DavidsonD. Role of the HPA axis and the A118G polymorphism of the mu-opioid receptor in stress-induced drinking behavior. Alcohol Alcohol.44(4) , 358–365 (2009).
  • Adinoff B , KrebaumSR, ChandlerPA, YeW, BrownMB, WilliamsMJ. Dissection of hypothalamic–pituitary–adrenal axis pathology in 1-month-abstinent alcohol-dependent men, part 2: response to ovine corticotropin-releasing factor and naloxone. Alcohol Clin. Exp. Res.29(4) , 528–537 (2005).
  • Katner SN , WeissF. Neurochemical characteristics associated with ethanol preference in selected alcohol-preferring and -nonpreferring rats: a quantitative microdialysis study. Alcohol Clin. Exp. Res.25(2) , 198–205 (2001).
  • Leyton M . Conditioned and sensitized responses to stimulant drugs in humans. Prog. Neuropsychopharmacol. Biol. Psychiatry31(8) , 1601–1613 (2007).
  • Gonzales RA , WeissF. Suppression of ethanol-reinforced behavior by naltrexone is associated with attenuation of the ethanol-induced increase in dialysate dopamine levels in the nucleus accumbens. J. Neurosci.18(24) , 10663–10671 (1998).
  • Stromberg MF , VolpicelliJR, O‘BrienCP. Effects of naltrexone administered repeatedly across 30 or 60 days on ethanol consumption using a limited access procedure in the rat. Alcohol Clin. Exp. Res.22(9) , 2186–2191 (1998).
  • Altshuler HL , PhillipsPE, FeinhandlerDA. Alteration of ethanol self-administration by naltrexone. Life Sci.26(9) , 679–688 (1980).
  • O‘Malley SS , Krishnan-SarinS, FarrenC, SinhaR, KreekMJ. Naltrexone decreases craving and alcohol self-administration in alcohol-dependent subjects and activates the hypothalamo-pituitary-adrenocortical axis. Psychopharmacology (Berl.)160(1) , 19–29 (2002).
  • Volpicelli JR , AltermanAI, HayashidaM, O‘BrienCP. Naltrexone in the treatment of alcohol dependence. Arch. Gen. Psychiatry49(11) , 876–880 (1992).
  • O‘Malley SS , JaffeAJ, ChangG, SchottenfeldRS, MeyerRE, RounsavilleB. Naltrexone and coping skills therapy for alcohol dependence. A controlled study. Arch. Gen. Psychiatry49(11) , 881–887 (1992).
  • Na C , LeeYS. Alcohol urge and plasma beta-endorphin change after alcohol challenge with naltrexone pretreatment in social drinkers. Prog. Neuropsychopharmacol. Biol. Psychiatry26(4) , 663–670 (2002).
  • Mccaul ME , WandGS, EissenbergT, RohdeCA, CheskinLJ. Naltrexone alters subjective and psychomotor responses to alcohol in heavy drinking subjects. Neuropsychopharmacology22(5) , 480–492 (2000).
  • Davidson D , PalfaiT, BirdC, SwiftR. Effects of naltrexone on alcohol self-administration in heavy drinkers. Alcohol Clin. Exp. Res.23(2) , 195–203 (1999).
  • Feinn R , KranzlerHR. Does effect size in naltrexone trials for alcohol dependence differ for single-site vs. multi-center studies? Alcohol Clin. Exp. Res.29(6) , 983–988 (2005).
  • Krystal JH , CramerJA, KrolWF, KirkGF, RosenheckRA. Naltrexone in the treatment of alcohol dependence. N. Engl. J. Med.345(24) , 1734–1739 (2001).
  • King AC , VolpicelliJR, FrazerA, O‘BrienCP. Effect of naltrexone on subjective alcohol response in subjects at high and low risk for future alcohol dependence. Psychopharmacology (Berl.)129(1) , 15–22 (1997).
  • Krishnan-Sarin S , KrystalJH, ShiJ, PittmanB, O‘MalleySS. Family history of alcoholism influences naltrexone-induced reduction in alcohol drinking. Biol. Psychiatry62(6) , 694–697 (2007).
  • Oslin DW , BerrettiniW, KranzlerHR et al. A functional polymorphism of the mu-opioid receptor gene is associated with naltrexone response in alcohol-dependent patients. Neuropsychopharmacology 28(8) , 1546–1552 (2003).
  • Anton RF , OrosziG, O‘MalleyS et al. An evaluation of mu-opioid receptor (OPRM1) as a predictor of naltrexone response in the treatment of alcohol dependence: results from the Combined Pharmacotherapies and Behavioral Interventions for Alcohol Dependence (COMBINE) study. Arch. Gen. Psychiatry 65(2) , 135–144 (2008).
  • Anton RF , O‘MalleySS, CirauloDA et al. Combined pharmacotherapies and behavioral interventions for alcohol dependence: the COMBINE study: a randomized controlled trial. JAMA 295(17) , 2003–2017 (2006).
  • Oroszi G , AntonRF, O‘MalleyS et al. OPRM1 Asn40Asp predicts response to naltrexone treatment: a haplotype-based approach. Alcohol Clin. Exp. Res.33(3) , 383–393 (2009).
  • Kim SG , KimCM, ChoiSW et al. A micro opioid receptor gene polymorphism (A118G) and naltrexone treatment response in adherent Korean alcohol-dependent patients. Psychopharmacology (Berl.) 201(4) , 611–618 (2009).
  • Setiawan E , PihlRO, CoxSM et al. The effect of naltrexone on alcohol‘s stimulant properties and self-administration behavior in social drinkers: influence of gender and genotype. Alcohol Clin. Exp. Res. 35(6) , 1134–1141 (2011).
  • Coller JK , CahillS, EdmondsC et al. OPRM1 A118G genotype fails to predict the effectiveness of naltrexone treatment for alcohol dependence. Pharmacogenet. Genomics21(12) , 902–905 (2011).
  • Gelernter J , GueorguievaR, KranzlerHR et al. Opioid receptor gene (OPRM1, OPRK1, and OPRD1) variants and response to naltrexone treatment for alcohol dependence: results from the VA cooperative study. Alcohol. Clin. Exp. Res. 31(4) , 555–563 (2007).
  • Tidey JW , MontiPM, RohsenowDJ et al. Moderators of naltrexone‘s effects on drinking, urge, and alcohol effects in non-treatment-seeking heavy drinkers in the natural environment. Alcohol. Clin. Exp. Res. 32(1) , 58–66 (2008).
  • Anton RF , VoroninKK, RandallPK, MyrickH, TiffanyA. Naltrexone modification of drinking effects in a subacute treatment and bar-lab paradigm: influence of OPRM1 and dopamine transporter (SLC6A3) genes. Alcohol. Clin. Exp. Res. doi:10.1111/j.1530-0277.2012.01807.x (2012) (Epub ahead of print).
  • Adlaf EM , BeginP, SawkaE (Eds). Canadian Addiction Survey (CAS): A National Survey of Canadians‘ Use of Alcohol and Other Drugs: Prevalence of Use and Related Harms: Detailed Report. Canadian Centre on Substance Abuse, Ottawa, Canada (2005).
  • Hasin DS , StinsonFS, OgburnE, GrantBF. Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States: results from the National Epidemiologic Survey on Alcohol and Related Conditions. Arch. Gen. Psychiatry64(7) , 830–842 (2007).
  • Urban NB , KegelesLS, SlifsteinM et al. Sex differences in striatal dopamine release in young adults after oral alcohol challenge: a positron emission tomography imaging study with [(1)(1)C]raclopride. Biol. Psychiatry 68(8) , 689–696 (2010).
  • Kiefer F , JahnH, WiedemannK. A neuroendocrinological hypothesis on gender effects of naltrexone in relapse prevention treatment. Pharmacopsychiatry38(4) , 184–186 (2005).
  • Job MO , TangA, HallFS et al. Mu (mu) opioid receptor regulation of ethanol-induced dopamine response in the ventral striatum: evidence of genotype specific sexual dimorphic epistasis. Biol. Psychiatry 62(6) , 627–634 (2007).
  • Lee MR , GallenCL, ZhangX et al. Functional polymorphism of the mu-opioid receptor gene (OPRM1) influences reinforcement learning in humans. PLoS ONE 6(9) , e24203 (2011).
  • Troisi A , FrazzettoG, CarolaV et al. Social hedonic capacity is associated with the A118G polymorphism of the mu-opioid receptor gene (OPRM1) in adult healthy volunteers and psychiatric patients. Soc. Neurosci. 6(1) , 88–97 (2011).
  • Ray R , JepsonC, PattersonF et al. Association of OPRM1 A118G variant with the relative reinforcing value of nicotine. Psychopharmacology (Berl.) 188(3) , 355–363 (2006).
  • Domino EF , EvansCL, NiL, GuthrieSK, KoeppeRA, ZubietaJK. Tobacco smoking produces greater striatal dopamine release in G-allele carriers with mu opioid receptor A118G polymorphism. Prog. Neuropsychopharmacol. Biol. Psychiatry doi:10.1016/j.pnpbp.2012.04.003 (2012) (Epub ahead of print).
  • Kleinjan M , PoelenEA, EngelsRC, VerhagenM. Dual growth of adolescent smoking and drinking: evidence for an interaction between the mu-opioid receptor (OPRM1) A118G polymorphism and sex. Addict Biol. doi:10.1111/j.1369-1600.2011.00422.x (2012) (Epub ahead of print).
  • Weerts EM , KimYK, WandGS et al. Differences in delta- and mu-opioid receptor blockade measured by positron emission tomography in naltrexone-treated recently abstinent alcohol-dependent subjects. Neuropsychopharmacology 33(3) , 653–665 (2008).
  • Marinelli PW , FunkD, HardingS, LiZ, JuzytschW, LeAD. Roles of opioid receptor subtypes in mediating alcohol-seeking induced by discrete cues and context. Eur. J. Neurosci.30(4) , 671–678 (2009).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.