Skip to main content

Advertisement

Log in

Association of the OPRM1 Variant rs1799971 (A118G) with Non-Specific Liability to Substance Dependence in a Collaborative de novo Meta-Analysis of European-Ancestry Cohorts

  • Original Research
  • Published:
Behavior Genetics Aims and scope Submit manuscript

Abstract

The mu1 opioid receptor gene, OPRM1, has long been a high-priority candidate for human genetic studies of addiction. Because of its potential functional significance, the non-synonymous variant rs1799971 (A118G, Asn40Asp) in OPRM1 has been extensively studied, yet its role in addiction has remained unclear, with conflicting association findings. To resolve the question of what effect, if any, rs1799971 has on substance dependence risk, we conducted collaborative meta-analyses of 25 datasets with over 28,000 European-ancestry subjects. We investigated non-specific risk for “general” substance dependence, comparing cases dependent on any substance to controls who were non-dependent on all assessed substances. We also examined five specific substance dependence diagnoses: DSM-IV alcohol, opioid, cannabis, and cocaine dependence, and nicotine dependence defined by the proxy of heavy/light smoking (cigarettes-per-day >20 vs. ≤10). The G allele showed a modest protective effect on general substance dependence (OR = 0.90, 95 % C.I. [0.83–0.97], p value = 0.0095, N = 16,908). We observed similar effects for each individual substance, although these were not statistically significant, likely because of reduced sample sizes. We conclude that rs1799971 contributes to mechanisms of addiction liability that are shared across different addictive substances. This project highlights the benefits of examining addictive behaviors collectively and the power of collaborative data sharing and meta-analyses.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  • American Psychiatric Association (2013) Diagnostic and Statistical Manual of Mental Disorders (5th Edition), Washington, DC

  • Andrews JA, Tildesley E, Hops H, Duncan SC, Severson HH (2003) Elementary school age children’s future intentions and use of substances. J Clin Child Adolesc Psychol 32(4):556–567

    Article  PubMed Central  PubMed  Google Scholar 

  • Arias A, Feinn R, Kranzler HR (2006) 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

    Article  PubMed  Google Scholar 

  • Aromaa A, Koskinen S (2004) Health and functional capacity in Finland: Baseline Results of the Health 2000 Health Examination Survey. National Public Health Institute, Helsinki

    Google Scholar 

  • Bart G, Kreek MJ, Ott J, LaForge KS, Proudnikov D, Pollak L, Heilig M (2005) 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

    Article  PubMed  Google Scholar 

  • Befort K, Filliol D, Decaillot FM, Gaveriaux-Ruff C, Hoehe MR, Kieffer BL (2001) A single nucleotide polymorphic mutation in the human mu-opioid receptor severely impairs receptor signaling. J Biol Chem 276(5):3130–3137

    Article  PubMed  Google Scholar 

  • Bergen AW, Kokoszka J, Peterson R, Long JC, Virkkunen M, Linnoila M, Goldman D (1997) Mu opioid receptor gene variants: lack of association with alcohol dependence. Mol Psychiatry 2(6):490–494

    Article  PubMed  Google Scholar 

  • Beyer A, Koch T, Schroder H, Schulz S, Hollt V (2004) 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

    Article  PubMed  Google Scholar 

  • Bierut LJ, Dinwiddie SH, Begleiter H, Crowe RR, Hesselbrock V, Nurnberger JI Jr, Porjesz B, Schuckit MA, Reich T (1998) Familial transmission of substance dependence: alcohol, marijuana, cocaine, and habitual smoking: a report from the Collaborative Study on the Genetics of Alcoholism. Arch Gen Psychiatry 55(11):982–988

    Article  PubMed  Google Scholar 

  • Bierut LJ, Strickland JR, Thompson JR, Afful SE, Cottler LB (2008) Drug use and dependence in cocaine dependent subjects, community-based individuals, and their siblings. Drug Alcohol Depend 95(1–2):14–22

    Article  PubMed Central  PubMed  Google Scholar 

  • Bierut LJ, Agrawal A, Bucholz KK, Doheny KF, Laurie C, Pugh E, Fisher S, Fox L, Howells W, Bertelsen S, Hinrichs AL, Almasy L, Breslau N, Culverhouse RC, Dick DM, Edenberg HJ, Foroud T, Grucza RA, Hatsukami D, Hesselbrock V, Johnson EO, Kramer J, Krueger RF, Kuperman S, Lynskey M, Mann K, Neuman RJ, Nothen MM, Nurnberger JI Jr, Porjesz B, Ridinger M, Saccone NL, Saccone SF, Schuckit MA, Tischfield JA, Wang JC, Rietschel M, Goate AM, Rice JP (2010) A genome-wide association study of alcohol dependence. Proc Natl Acad Sci USA 107(11):5082–5087

    Article  PubMed Central  PubMed  Google Scholar 

  • Bond C, LaForge KS, Tian M, Melia D, Zhang S, Borg L, Gong J, Schluger J, Strong JA, Leal SM, Tischfield JA, Kreek MJ, Yu L (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 USA 95(16):9608–9613

    Article  PubMed Central  PubMed  Google Scholar 

  • Broms U, Wedenoja J, Largeau MR, Korhonen T, Pitkaniemi J, Keskitalo-Vuokko K, Happola A, Heikkila KH, Heikkila K, Ripatti S, Sarin AP, Salminen O, Paunio T, Pergadia ML, Madden PA, Kaprio J, Loukola A (2012) Analysis of detailed phenotype profiles reveals CHRNA5-CHRNA3-CHRNB4 gene cluster association with several nicotine dependence traits. Nicotine Tob Res 14(6):720–733

    Article  PubMed Central  PubMed  Google Scholar 

  • Carter BL, Lam CY, Robinson JD, Paris MM, Waters AJ, Wetter DW, Cinciripini PM (2008) Real-time craving and mood assessments before and after smoking. Nicotine Tob Res 10(7):1165–1169

    Article  PubMed Central  PubMed  Google Scholar 

  • Chen X, Chen J, Williamson VS, An SS, Hettema JM, Aggen SH, Neale MC, Kendler KS (2009) Variants in nicotinic acetylcholine receptors alpha5 and alpha3 increase risks to nicotine dependence. Am J Med Genet B Neuropsychiatr Genet

  • Chen D, Liu L, Xiao Y, Peng Y, Yang C, Wang Z (2012a) 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

    Article  PubMed  Google Scholar 

  • Chen LS, Saccone NL, Culverhouse RC, Bracci PM, Chen CH, Dueker N, Han Y, Huang H, Jin G, Kohno T, Ma JZ, Przybeck TR, Sanders AR, Smith JA, Sung YJ, Wenzlaff AS, Wu C, Yoon D, Chen YT, Cheng YC, Cho YS, David SP, Duan J, Eaton CB, Furberg H, Goate AM, Gu D, Hansen HM, Hartz S, Hu Z, Kim YJ, Kittner SJ, Levinson DF, Mosley TH, Payne TJ, Rao DC, Rice JP, Rice TK, Schwantes-An TH, Shete SS, Shi J, Spitz MR, Sun YV, Tsai FJ, Wang JC, Wrensch MR, Xian H, Gejman PV, He J, Hunt SC, Kardia SL, Li MD, Lin D, Mitchell BD, Park T, Schwartz AG, Shen H, Wiencke JK, Wu JY, Yokota J, Amos CI, Bierut LJ (2012b) Smoking and genetic risk variation across populations of European, Asian, and African American ancestry–a meta-analysis of chromosome 15q25. Genet Epidemiol 36(4):340–351

    Article  PubMed Central  PubMed  Google Scholar 

  • Cheverud JM (2001) A simple correction for multiple comparisons in interval mapping genome scans. Heredity 87(Pt 1):52–58

    Article  PubMed  Google Scholar 

  • Cinciripini PM, Tsoh JY, Wetter DW, Lam C, de Moor C, Cinciripini L, Baile W, Anderson C, Minna JD (2005) Combined effects of venlafaxine, nicotine replacement, and brief counseling on smoking cessation. Exp Clin Psychopharmacol 13(4):282–292

    Article  PubMed  Google Scholar 

  • Cinciripini PM, Robinson JD, Carter BL, Lam C, Wu X, de Moor CA, Baile WF, Wetter DW (2006) The effects of smoking deprivation and nicotine administration on emotional reactivity. Nicotine Tob Res 8(3):379–392

    Article  PubMed  Google Scholar 

  • Clarke TK, Crist RC, Kampman KM, Dackis CA, Pettinati HM, O’Brien CP, Oslin DW, Ferraro TN, Lohoff FW, Berrettini WH (2013) Low frequency genetic variants in the mu-opioid receptor (OPRM1) affect risk for addiction to heroin and cocaine. Neurosci Lett 54271-75

  • Coller JK, Beardsley J, Bignold J, Li Y, Merg F, Sullivan T, Cox TC, Somogyi AA (2009) Lack of association between the A118G polymorphism of the mu opioid receptor gene (OPRM1) and opioid dependence: A meta-analysis. Pharmgenomics Pers Med 29-19

  • Crist RC, Berrettini WH (2013) Pharmacogenetics of OPRM1. Pharmacol Biochem Behav

  • Cross-Disorder Group of the Psychiatric Genomics Consortium (2013) Identification of risk loci with shared effects on five major psychiatric disorders: a genome-wide analysis. Lancet 381(9875):1371–1379

    Article  PubMed Central  Google Scholar 

  • Crowley JJ, Oslin DW, Patkar AA, Gottheil E, DeMaria PA Jr, O’Brien CP, Berrettini WH, Grice DE (2003) A genetic association study of the mu opioid receptor and severe opioid dependence. Psychiatr Genet 13(3):169–173

    Article  PubMed  Google Scholar 

  • David SP, Johnstone EC, Churchman M, Aveyard P, Murphy MF, Munafo MR (2011) Pharmacogenetics of smoking cessation in general practice: results from the Patch II and Patch in Practice trials. Nicotine Tob Res 13(3):157–167

    Article  PubMed Central  PubMed  Google Scholar 

  • Deb I, Chakraborty J, Gangopadhyay PK, Choudhury SR, Das S (2010) Single-nucleotide polymorphism (A118G) in exon 1 of OPRM1 gene causes alteration in downstream signaling by mu-opioid receptor and may contribute to the genetic risk for addiction. J Neurochem 112(2):486–496

    Article  PubMed  Google Scholar 

  • Development Core Team R (2012) R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna

    Google Scholar 

  • Drakenberg K, Nikoshkov A, Horvath MC, Fagergren P, Gharibyan A, Saarelainen K, Rahman S, Nylander I, Bakalkin G, Rajs J, Keller E, Hurd YL (2006) Mu opioid receptor A118G polymorphism in association with striatal opioid neuropeptide gene expression in heroin abusers. Proc Natl Acad Sci USA 103(20):7883–7888

    Article  PubMed Central  PubMed  Google Scholar 

  • Edenberg HJ (2002) The collaborative study on the genetics of alcoholism: an update. Alcohol Res Health 26(3):214–218

    PubMed  Google Scholar 

  • Edenberg HJ, Koller DL, Xuei X, Wetherill L, McClintick JN, Almasy L, Bierut LJ, Bucholz KK, Goate A, Aliev F, Dick D, Hesselbrock V, Hinrichs A, Kramer J, Kuperman S, Nurnberger JI Jr, Rice JP, Schuckit MA, Taylor R, Todd Webb B, Tischfield JA, Porjesz B, Foroud T (2010) Genome-wide association study of alcohol dependence implicates a region on chromosome 11. Alcohol Clin Exp Res 34(5):840–852

    Article  PubMed Central  PubMed  Google Scholar 

  • Elliott DS, Huizinga D, Ageton SS (1985) Explaining delinquency and drug use. Sage Publications, Beverly Hills

    Google Scholar 

  • Elliott DS, Huizinga D, Menard S (1989) Multiple Problem Youth: Delinquency. Drugs and Mental Health Problems, Springer

    Book  Google Scholar 

  • Frank J, Cichon S, Treutlein J, Ridinger M, Mattheisen M, Hoffmann P, Herms S, Wodarz N, Soyka M, Zill P, Maier W, Mossner R, Gaebel W, Dahmen N, Scherbaum N, Schmal C, Steffens M, Lucae S, Ising M, Muller-Myhsok B, Nothen MM, Mann K, Kiefer F, Rietschel M (2012) Genome-wide significant association between alcohol dependence and a variant in the ADH gene cluster. Addict Biol 17(1):171–180

    Article  PubMed Central  PubMed  Google Scholar 

  • Franke P, Wang T, Nothen MM, Knapp M, Neidt H, Albrecht S, Jahnes E, Propping P, Maier W (2001) Nonreplication of association between mu-opioid-receptor gene (OPRM1) A118G polymorphism and substance dependence. Am J Med Genet 105(1):114–119

    Article  PubMed  Google Scholar 

  • Gelernter J, Kranzler H, Cubells J (1999) Genetics of two mu opioid receptor gene (OPRM1) exon I polymorphisms: population studies, and allele frequencies in alcohol- and drug-dependent subjects. Mol Psychiatry 4(5):476–483

    Article  PubMed  Google Scholar 

  • Gelernter J, Kranzler HR, Sherva R, Koesterer R, Almasy L, Zhao H, Farrer LA (2013a) Genome-Wide association study of opioid dependence: multiple associations mapped to calcium and potassium pathways. Biol Psychiatry

  • Gelernter J, Sherva R, Koesterer R, Almasy L, Zhao H, Kranzler HR, Farrer L (2013b) Genome-wide association study of cocaine dependence and related traits: FAM53B identified as a risk gene. Mol Psychiatry

  • Gelernter J, Kranzler HR, Sherva R, Almasy L, Koesterer R, Smith AH, Anton R, Preuss UW, Ridinger M, Rujescu D, Wodarz N, Zill P, Zhao H, Farrer LA (2014) Genome-wide association study of alcohol dependence: significant findings in African- and European-Americans including novel risk loci. Mol Psychiatry 19(1):41–49

    Article  PubMed Central  PubMed  Google Scholar 

  • Glatt SJ, Bousman C, Wang RS, Murthy KK, Rana BK, Lasky-Su JA, Zhu SC, Zhang R, Li J, Zhang B, Lyons MJ, Faraone SV, Tsuang MT (2007) Evaluation of OPRM1 variants in heroin dependence by family-based association testing and meta-analysis. Drug Alcohol Depend 90(2–3):159–165

    Article  PubMed Central  PubMed  Google Scholar 

  • Grucza RA, Wang JC, Stitzel JA, Hinrichs AL, Saccone SF, Saccone NL, Bucholz KK, Cloninger CR, Neuman RJ, Budde JP, Fox L, Bertelsen S, Kramer J, Hesselbrock V, Tischfield J, Nurnberger JI Jr, Almasy L, Porjesz B, Kuperman S, Schuckit MA, Edenberg HJ, Rice JP, Goate AM, Bierut LJ (2008) A risk allele for nicotine dependence in CHRNA5 is a protective allele for cocaine dependence. Biol Psychiatry 64(11):922–929

    Article  PubMed Central  PubMed  Google Scholar 

  • Haerian BS, Haerian MS (2013) OPRM1 rs1799971 polymorphism and opioid dependence: evidence from a meta-analysis. Pharmacogenomics 14(7):813–824

    Article  PubMed  Google Scholar 

  • Hall FS, Drgonova J, Jain S, Uhl GR (2013) Implications of genome wide association studies for addiction: are our a priori assumptions all wrong? Pharmacol Ther 140(3):267–279

    Article  PubMed Central  PubMed  Google Scholar 

  • Hancock DB, Levy JL, Gaddis NC, Glasheen C, Saccone NL, Page GP, Hulse GK, Wildenauer D, Kelty EA, Schwab SG, Degenhardt L, Martin NG, Montgomery GW, Attia J, Holliday EG, McEvoy M, Scott RJ, Bierut LJ, Nelson EC, Kral AH, Johnson EO (2015) Cis-Expression Quantitative Trait Loci Mapping Reveals Replicable Associations with Heroin Addiction in OPRM1. Biol Psychiatry

  • Hardin J, He Y, Javitz HS, Wessel J, Krasnow RE, Tildesley E, Hops H, Swan GE, Bergen AW (2009) Nicotine withdrawal sensitivity, linkage to chr6q26, and association of OPRM1 SNPs in the SMOking in FAMilies (SMOFAM) sample. Cancer Epidemiol Biomarkers Prev 18(12):3399–3406

    Article  PubMed Central  PubMed  Google Scholar 

  • Hart AB, de Wit H, Palmer AA (2013) Candidate gene studies of a promising intermediate phenotype: failure to replicate. Neuropsychopharmacology 38(5):802–816

    Article  PubMed Central  PubMed  Google Scholar 

  • Hartz SM, Short SE, Saccone NL, Culverhouse R, Chen L, Schwantes-An TH, Coon H, Han Y, Stephens SH, Sun J, Chen X, Ducci F, Dueker N, Franceschini N, Frank J, Geller F, Gubjartsson D, Hansel NN, Jiang C, Keskitalo-Vuokko K, Liu Z, Lyytikainen LP, Michel M, Rawal R, Rosenberger A, Scheet P, Shaffer JR, Teumer A, Thompson JR, Vink JM, Vogelzangs N, Wenzlaff AS, Wheeler W, Xiao X, Yang BZ, Aggen SH, Balmforth AJ, Baumeister SE, Beaty T, Bennett S, Bergen AW, Boyd HA, Broms U, Campbell H, Chatterjee N, Chen J, Cheng YC, Cichon S, Couper D, Cucca F, Dick DM, Foroud T, Furberg H, Giegling I, Gu F, Hall AS, Hallfors J, Han S, Hartmann AM, Hayward C, Heikkila K, Hewitt JK, Hottenga JJ, Jensen MK, Jousilahti P, Kaakinen M, Kittner SJ, Konte B, Korhonen T, Landi MT, Laatikainen T, Leppert M, Levy SM, Mathias RA, McNeil DW, Medland SE, Montgomery GW, Muley T, Murray T, Nauck M, North K, Pergadia M, Polasek O, Ramos EM, Ripatti S, Risch A, Ruczinski I, Rudan I, Salomaa V, Schlessinger D, Styrkarsdottir U, Terracciano A, Uda M, Willemsen G, Wu X, Abecasis G, Barnes K, Bickeboller H, Boerwinkle E, Boomsma DI, Caporaso N, Duan J, Edenberg HJ, Francks C, Gejman PV, Gelernter J, Grabe HJ, Hops H, Jarvelin MR, Viikari J, Kahonen M, Kendler KS, Lehtimaki T, Levinson DF, Marazita ML, Marchini J, Melbye M, Mitchell BD, Murray JC, Nothen MM, Penninx BW, Raitakari O, Rietschel M, Rujescu D, Samani NJ, Sanders AR, Schwartz AG, Shete S, Shi J, Spitz M, Stefansson K, Swan GE, Thorgeirsson T, Volzke H, Wei Q, Wichmann HE, Amos CI, Breslau N, Cannon DS, Ehringer M, Grucza R, Hatsukami D, Heath A, Johnson EO, Kaprio J, Madden P, Martin NG, Stevens VL, Stitzel JA, Weiss RB, Kraft P, Bierut LJ (2012) Increased Genetic Vulnerability to Smoking at CHRNA5 in Early-Onset Smokers. Arch Gen Psychiatry 69(8):854–860

    Article  PubMed Central  PubMed  Google Scholar 

  • Heatherton TF, Kozlowski LT, Frecker RC, Fagerstrom KO (1991) The Fagerstrom Test for Nicotine Dependence: a revision of the Fagerstrom Tolerance Questionnaire. Br J Addict 86(9):1119–1127

    Article  PubMed  Google Scholar 

  • Hoft NR, Corley RP, McQueen MB, Schlaepfer IR, Huizinga D, Ehringer MA (2009) Genetic association of the CHRNA6 and CHRNB3 genes with tobacco dependence in a nationally representative sample. Neuropsychopharmacology 34(3):698–706

    Article  PubMed Central  PubMed  Google Scholar 

  • Hops H, Andrews JA, Duncan SC, Duncan TE, Tildesley E (2000) Adolescent drug use development: a social interactional and contextual perspective. In: Sameroff AJ, Lewis M (eds) Handbook of Developmental Psychopathology. Cluwer Academic/Plenum, New York, pp 589–605

    Chapter  Google Scholar 

  • Huang P, Chen C, Mague SD, Blendy JA, Liu-Chen LY (2012) A common single nucleotide polymorphism A118G of the mu opioid receptor alters its N-glycosylation and protein stability. Biochem J 441(1):379–386

    Article  PubMed Central  PubMed  Google Scholar 

  • Iacono WG, Carlson SR, Taylor J, Elkins IJ, McGue M (1999) Behavioral disinhibition and the development of substance-use disorders: findings from the Minnesota Twin Family Study. Dev Psychopathol 11(4):869–900

    Article  PubMed  Google Scholar 

  • Kamens HM, Corley RP, McQueen MB, Stallings MC, Hopfer CJ, Crowley TJ, Brown SA, Hewitt JK, Ehringer MA (2013) Nominal association with CHRNA4 variants and nicotine dependence. Genes Brain Behav 12(3):297–304

    Article  PubMed  Google Scholar 

  • Kaprio J, Pulkkinen L, Rose RJ (2002) Genetic and environmental factors in health-related behaviors: studies on Finnish twins and twin families. Twin Res 5(5):366–371

    Article  PubMed  Google Scholar 

  • Kendler KS, Myers J, Prescott CA (2007) Specificity of genetic and environmental risk factors for symptoms of cannabis, cocaine, alcohol, caffeine, and nicotine dependence. Arch Gen Psychiatry 64(11):1313–1320

    Article  PubMed  Google Scholar 

  • Keyes MA, Malone SM, Elkins IJ, Legrand LN, McGue M, Iacono WG (2009) The enrichment study of the Minnesota twin family study: increasing the yield of twin families at high risk for externalizing psychopathology. Twin Res Hum Genet 12(5):489–501

    Article  PubMed Central  PubMed  Google Scholar 

  • Kim SG, Kim CM, Kang DH, Kim YJ, Byun WT, Kim SY, Park JM, Kim MJ, Oslin DW (2004) Association of functional opioid receptor genotypes with alcohol dependence in Koreans. Alcohol Clin Exp Res 28(7):986–990

    Article  PubMed  Google Scholar 

  • Kreek MJ, Koob GF (1998) Drug dependence: stress and dysregulation of brain reward pathways. Drug Alcohol Depend 51(1–2):23–47

    Article  PubMed  Google Scholar 

  • Lam CY, Robinson JD, Versace F, Minnix JA, Cui Y, Carter BL, Wetter DW, Cinciripini PM (2012) Affective reactivity during smoking cessation of never-quitters as compared with that of abstainers, relapsers, and continuing smokers. Exp Clin Psychopharmacol 20(2):139–150

    Article  PubMed Central  PubMed  Google Scholar 

  • Lessov CN, Martin NG, Statham DJ, Todorov AA, Slutske WS, Bucholz KK, Heath AC, Madden PA (2004) Defining nicotine dependence for genetic research: evidence from Australian twins. Psychol Med 34(5):865–879

    Article  PubMed  Google Scholar 

  • Levran O, Londono D, O’Hara K, Nielsen DA, Peles E, Rotrosen J, Casadonte P, Linzy S, Randesi M, Ott J, Adelson M, Kreek MJ (2008) Genetic susceptibility to heroin addiction: a candidate gene association study. Genes Brain Behav 7(7):720–729

    Article  PubMed Central  PubMed  Google Scholar 

  • Levran O, Yuferov V, Kreek MJ (2012) The genetics of the opioid system and specific drug addictions. Hum Genet 131(6):823–842

    Article  PubMed Central  PubMed  Google Scholar 

  • Li J, Ji L (2005) Adjusting multiple testing in multilocus analyses using the eigenvalues of a correlation matrix. Heredity 95(3):221–227

    Article  PubMed  Google Scholar 

  • Liu JZ, Tozzi F, Waterworth DM, Pillai SG, Muglia P, Middleton L, Berrettini W, Knouff CW, Yuan X, Waeber G, Vollenweider P, Preisig M, Wareham NJ, Zhao JH, Loos RJ, Barroso I, Khaw KT, Grundy S, Barter P, Mahley R, Kesaniemi A, McPherson R, Vincent JB, Strauss J, Kennedy JL, Farmer A, McGuffin P, Day R, Matthews K, Bakke P, Gulsvik A, Lucae S, Ising M, Brueckl T, Horstmann S, Wichmann HE, Rawal R, Dahmen N, Lamina C, Polasek O, Zgaga L, Huffman J, Campbell S, Kooner J, Chambers JC, Burnett MS, Devaney JM, Pichard AD, Kent KM, Satler L, Lindsay JM, Waksman R, Epstein S, Wilson JF, Wild SH, Campbell H, Vitart V, Reilly MP, Li M, Qu L, Wilensky R, Matthai W, Hakonarson HH, Rader DJ, Franke A, Wittig M, Schafer A, Uda M, Terracciano A, Xiao X, Busonero F, Scheet P, Schlessinger D, St Clair D, Rujescu D, Abecasis GR, Grabe HJ, Teumer A, Volzke H, Petersmann A, John U, Rudan I, Hayward C, Wright AF, Kolcic I, Wright BJ, Thompson JR, Balmforth AJ, Hall AS, Samani NJ, Anderson CA, Ahmad T, Mathew CG, Parkes M, Satsangi J, Caulfield M, Munroe PB, Farrall M, Dominiczak A, Worthington J, Thomson W, Eyre S, Barton A, Mooser V, Francks C, Marchini J (2010) Meta-analysis and imputation refines the association of 15q25 with smoking quantity. Nat Genet 42(5):436–440

    Article  PubMed Central  PubMed  Google Scholar 

  • Loukola A, Broms U, Maunu H, Widen E, Heikkila K, Siivola M, Salo A, Pergadia ML, Nyman E, Sammalisto S, Perola M, Agrawal A, Heath AC, Martin NG, Madden PA, Peltonen L, Kaprio J (2008) Linkage of nicotine dependence and smoking behavior on 10q, 7q and 11p in twins with homogeneous genetic background. Pharmacogenomics J 8(3):209–219

    Article  PubMed  Google Scholar 

  • Lumley T (2009) rmeta: Meta-analysis, R package version 2.15. http://CRAN.R-project.org/package=rmeta

  • Mague SD, Blendy JA (2010) OPRM1 SNP (A118G): involvement in disease development, treatment response, and animal models. Drug Alcohol Depend 108(3):172–182

    Article  PubMed Central  PubMed  Google Scholar 

  • Mague SD, Isiegas C, Huang P, Liu-Chen LY, Lerman C, Blendy JA (2009) Mouse model of OPRM1 (A118G) polymorphism has sex-specific effects on drug-mediated behavior. Proc Natl Acad Sci USA 106(26):10847–10852

    Article  PubMed Central  PubMed  Google Scholar 

  • Maher BS, Vladimirov VI, Latendresse SJ, Thiselton DL, McNamee R, Kang M, Bigdeli TB, Chen X, Riley BP, Hettema JM, Chilcoat H, Heidbreder C, Muglia P, Murrelle EL, Dick DM, Aliev F, Agrawal A, Edenberg HJ, Kramer J, Nurnberger J, Tischfield JA, Devlin B, Ferrell RE, Kirillova GP, Tarter RE, Kendler KS, Vanyukov MM (2011) The AVPR1A gene and substance use disorders: association, replication, and functional evidence. Biol Psychiatry 70(6):519–527

    Article  PubMed Central  PubMed  Google Scholar 

  • McGue M, Keyes M, Sharma A, Elkins I, Legrand L, Johnson W, Iacono WG (2007) The environments of adopted and non-adopted youth: evidence on range restriction from the Sibling Interaction and Behavior Study (SIBS). Behav Genet 37(3):449–462

    Article  PubMed  Google Scholar 

  • Medland SE, Neale MC (2010) An integrated phenomic approach to multivariate allelic association. Eur J Hum Genet 18(2):233–239

    Article  PubMed Central  PubMed  Google Scholar 

  • Merikangas KR, Stolar M, Stevens DE, Goulet J, Preisig MA, Fenton B, Zhang H, O’Malley SS, Rounsaville BJ (1998) Familial transmission of substance use disorders. Arch Gen Psychiatry 55(11):973–979

    Article  PubMed  Google Scholar 

  • Miller MB, Basu S, Cunningham J, Eskin E, Malone SM, Oetting WS, Schork N, Sul JH, Iacono WG, McGue M (2012) The Minnesota Center for Twin and Family Research genome-wide association study. Twin Res Hum Genet 15(6):767–774

    Article  PubMed Central  PubMed  Google Scholar 

  • Minnix JA, Robinson JD, Lam CY, Carter BL, Foreman JE, Vandenbergh DJ, Tomlinson GE, Wetter DW, Cinciripini PM (2011) The serotonin transporter gene and startle response during nicotine deprivation. Biol Psychol 86(1):1–8

    Article  PubMed Central  PubMed  Google Scholar 

  • Miranda R Jr, Reynolds E, Ray L, Justus A, Knopik VS, McGeary J, Meyerson LA (2013) Preliminary evidence for a gene-environment interaction in predicting alcohol use disorders in adolescents. Alcohol Clin Exp Res 37(2):325–331

    Article  PubMed Central  PubMed  Google Scholar 

  • Munafo MR, Johnstone EC, Aveyard P, Marteau T (2013) Lack of association of OPRM1 genotype and smoking cessation. Nicotine Tob Res 15(3):739–744

    Article  PubMed  Google Scholar 

  • Neale MC, Harvey E, Maes HH, Sullivan PF, Kendler KS (2006) Extensions to the modeling of initiation and progression: applications to substance use and abuse. Behav Genet 36(4):507–524

    Article  PubMed  Google Scholar 

  • Nelson EC, Lynskey MT, Heath AC, Wray N, Agrawal A, Shand FL, Henders AK, Wallace L, Todorov AA, Schrage AJ, Saccone NL, Madden PA, Degenhardt L, Martin NG, Montgomery GW (2013) ANKK1, TTC12, and NCAM1 polymorphisms and heroin dependence: importance of considering drug exposure. JAMA Psychiatry 70(3):325–333

    Article  PubMed Central  PubMed  Google Scholar 

  • Nelson EC, Lynskey MT, Heath AC, Wray N, Agrawal A, Shand FL, Henders AK, Wallace L, Todorov AA, Schrage AJ, Madden PA, Degenhardt L, Martin NG, Montgomery GW (2014) Association of OPRD1 polymorphisms with heroin dependence in a large case-control series. Addict Biol 19(1):111–121

    Article  PubMed  Google Scholar 

  • Nikolov MA, Beltcheva O, Galabova A, Ljubenova A, Jankova E, Gergov G, Russev AA, Lynskey MT, Nelson EC, Nesheva E, Krasteva D, Lazarov P, Mitev VI, Kremensky IM, Kaneva RP, Todorov AA (2011) No evidence of association between 118A > G OPRM1 polymorphism and heroin dependence in a large Bulgarian case-control sample. Drug Alcohol Depend 117(1):62–65

    Article  PubMed Central  PubMed  Google Scholar 

  • Nyholt DR (2004) A simple correction for multiple testing for single-nucleotide polymorphisms in linkage disequilibrium with each other. Am J Hum Genet 74(4):765–769

    Article  PubMed Central  PubMed  Google Scholar 

  • Palmer AA, de Wit H (2012) Translational genetic approaches to substance use disorders: bridging the gap between mice and humans. Hum Genet 131931-939

  • Ramchandani VA, Umhau J, Pavon FJ, Ruiz-Velasco V, Margas W, Sun H, Damadzic R, Eskay R, Schoor M, Thorsell A, Schwandt ML, Sommer WH, George DT, Parsons LH, Herscovitch P, Hommer D, Heilig M (2011) A genetic determinant of the striatal dopamine response to alcohol in men. Mol Psychiatry 16(8):809–817

    Article  PubMed Central  PubMed  Google Scholar 

  • Ray LA, Hutchison KE (2004) 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

    Article  PubMed  Google Scholar 

  • Ray LA, Hutchison KE (2007) Effects of naltrexone on alcohol sensitivity and genetic moderators of medication response: a double-blind placebo-controlled study. Arch Gen Psychiatry 64(9):1069–1077

    Article  PubMed  Google Scholar 

  • Ray R, Jepson C, Patterson F, Strasser A, Rukstalis M, Perkins K, Lynch KG, O’Malley S, Berrettini WH, Lerman C (2006) Association of OPRM1 A118G variant with the relative reinforcing value of nicotine. Psychopharmacology 188(3):355–363

    Article  PubMed  Google Scholar 

  • Ray LA, Miranda R Jr, Tidey JW, McGeary JE, MacKillop J, Gwaltney CJ, Rohsenow DJ, Swift RM, Monti PM (2010) 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

    Article  PubMed Central  PubMed  Google Scholar 

  • Ray R, Ruparel K, Newberg A, Wileyto EP, Loughead JW, Divgi C, Blendy JA, Logan J, Zubieta JK, Lerman C (2011) Human Mu Opioid Receptor (OPRM1 A118G) polymorphism is associated with brain mu-opioid receptor binding potential in smokers. Proc Natl Acad Sci USA 108(22):9268–9273

    Article  PubMed Central  PubMed  Google Scholar 

  • Ray LA, Barr CS, Blendy JA, Oslin D, Goldman D, Anton RF (2012) The role of the Asn40Asp polymorphism of the mu opioid receptor gene (OPRM1) on alcoholism etiology and treatment: a critical review. Alcohol Clin Exp Res 36(3):385–394

    Article  PubMed Central  PubMed  Google Scholar 

  • Robinson JD, Cinciripini PM, Carter BL, Lam CY, Wetter DW (2007) Facial EMG as an index of affective response to nicotine. Exp Clin Psychopharmacol 15(4):390–399

    Article  PubMed  Google Scholar 

  • Robinson JD, Lam CY, Carter BL, Minnix JA, Cui Y, Versace F, Wetter DW, Cinciripini PM (2011) A multimodal approach to assessing the impact of nicotine dependence, nicotine abstinence, and craving on negative affect in smokers. Exp Clin Psychopharmacol 19(1):40–52

    Article  PubMed Central  PubMed  Google Scholar 

  • Rouvinen-Lagerstrom N, Lahti J, Alho H, Kovanen L, Aalto M, Partonen T, Silander K, Sinclair D, Raikkonen K, Eriksson JG, Palotie A, Koskinen S, Saarikoski ST (2013) mu-Opioid receptor gene (OPRM1) polymorphism A118G: lack of association in Finnish populations with alcohol dependence or alcohol consumption. Alcohol Alcohol 48(5):519–525

    Article  PubMed Central  PubMed  Google Scholar 

  • Saccone SF, Pergadia ML, Loukola A, Broms U, Montgomery GW, Wang JC, Agrawal A, Dick DM, Heath AC, Todorov AA, Maunu H, Heikkila K, Morley KI, Rice JP, Todd RD, Kaprio J, Peltonen L, Martin NG, Goate AM, Madden PA (2007) Genetic linkage to chromosome 22q12 for a heavy-smoking quantitative trait in two independent samples. Am J Hum Genet 80(5):856–866

    Article  PubMed Central  PubMed  Google Scholar 

  • Saccone NL, Saccone SF, Hinrichs AL, Stitzel JA, Duan W, Pergadia ML, Agrawal A, Breslau N, Grucza RA, Hatsukami D, Johnson EO, Madden PA, Swan GE, Wang JC, Goate AM, Rice JP, Bierut LJ (2009a) Multiple distinct risk loci for nicotine dependence identified by dense coverage of the complete family of nicotinic receptor subunit (CHRN) genes. Am J Med Genet B Neuropsychiatr Genet 150B(4):453–466

    Article  PubMed Central  PubMed  Google Scholar 

  • Saccone NL, Wang JC, Breslau N, Johnson EO, Hatsukami D, Saccone SF, Grucza RA, Sun L, Duan W, Budde J, Culverhouse RC, Fox L, Hinrichs AL, Steinbach JH, Wu M, Rice JP, Goate AM, Bierut LJ (2009b) The CHRNA5-CHRNA3-CHRNB4 nicotinic receptor subunit gene cluster affects risk for nicotine dependence in African-Americans and in European-Americans. Cancer Res 69(17):6848–6856

    Article  PubMed Central  PubMed  Google Scholar 

  • Saccone NL, Culverhouse RC, Schwantes-An TH, Cannon DS, Chen X, Cichon S, Giegling I, Han S, Han Y, Keskitalo-Vuokko K, Kong X, Landi MT, Ma JZ, Short SE, Stephens SH, Stevens VL, Sun L, Wang Y, Wenzlaff AS, Aggen SH, Breslau N, Broderick P, Chatterjee N, Chen J, Heath AC, Heliovaara M, Hoft NR, Hunter DJ, Jensen MK, Martin NG, Montgomery GW, Niu T, Payne TJ, Peltonen L, Pergadia ML, Rice JP, Sherva R, Spitz MR, Sun J, Wang JC, Weiss RB, Wheeler W, Witt SH, Yang BZ, Caporaso NE, Ehringer MA, Eisen T, Gapstur SM, Gelernter J, Houlston R, Kaprio J, Kendler KS, Kraft P, Leppert MF, Li MD, Madden PA, Nothen MM, Pillai S, Rietschel M, Rujescu D, Schwartz A, Amos CI, Bierut LJ (2010) Multiple Independent Loci at Chromosome 15q25.1 Affect Smoking Quantity: a Meta-Analysis and Comparison with Lung Cancer and COPD. PLoS Genet 6(8):e1001053

    Article  PubMed Central  PubMed  Google Scholar 

  • Sander T, Gscheidel N, Wendel B, Samochowiec J, Smolka M, Rommelspacher H, Schmidt LG, Hoehe MR (1998) Human mu-opioid receptor variation and alcohol dependence. Alcohol Clin Exp Res 22(9):2108–2110

    PubMed  Google Scholar 

  • Schinka JA, Town T, Abdullah L, Crawford FC, Ordorica PI, Francis E, Hughes P, Graves AB, Mortimer JA, Mullan M (2002) A functional polymorphism within the mu-opioid receptor gene and risk for abuse of alcohol and other substances. Mol Psychiatry 7(2):224–228

    Article  PubMed  Google Scholar 

  • Stallings MC, Corley RP, Hewitt JK, Krauter KS, Lessem JM, Mikulich SK, Rhee SH, Smolen A, Young SE, Crowley TJ (2003) A genome-wide search for quantitative trait loci influencing substance dependence vulnerability in adolescence. Drug Alcohol Depend 70(3):295–307

    Article  PubMed  Google Scholar 

  • Stallings MC, Corley RP, Dennehey B, Hewitt JK, Krauter KS, Lessem JM, Mikulich-Gilbertson SK, Rhee SH, Smolen A, Young SE, Crowley TJ (2005) A genome-wide search for quantitative trait loci that influence antisocial drug dependence in adolescence. Arch Gen Psychiatry 62(9):1042–1051

    Article  PubMed  Google Scholar 

  • Swan GE, Carmelli D, Cardon LR (1997) Heavy consumption of cigarettes, alcohol and coffee in male twins. J Stud Alcohol 58(2):182–190

    Article  PubMed  Google Scholar 

  • Tan EC, Tan CH, Karupathivan U, Yap EP (2003) Mu opioid receptor gene polymorphisms and heroin dependence in Asian populations. NeuroReport 14(4):569–572

    Article  PubMed  Google Scholar 

  • The Tobacco and Genetics Consortium (2010) Genome-wide meta-analyses identify multiple loci associated with smoking behavior. Nat Genet 42(5):441–447

    Article  PubMed Central  Google Scholar 

  • Thorgeirsson TE, Gudbjartsson DF, Surakka I, Vink JM, Amin N, Geller F, Sulem P, Rafnar T, Esko T, Walter S, Gieger C, Rawal R, Mangino M, Prokopenko I, Magi R, Keskitalo K, Gudjonsdottir IH, Gretarsdottir S, Stefansson H, Thompson JR, Aulchenko YS, Nelis M, Aben KK, den Heijer M, Dirksen A, Ashraf H, Soranzo N, Valdes AM, Steves C, Uitterlinden AG, Hofman A, Tonjes A, Kovacs P, Hottenga JJ, Willemsen G, Vogelzangs N, Doring A, Dahmen N, Nitz B, Pergadia ML, Saez B, De Diego V, Lezcano V, Garcia-Prats MD, Ripatti S, Perola M, Kettunen J, Hartikainen AL, Pouta A, Laitinen J, Isohanni M, Huei-Yi S, Allen M, Krestyaninova M, Hall AS, Jones GT, van Rij AM, Mueller T, Dieplinger B, Haltmayer M, Jonsson S, Matthiasson SE, Oskarsson H, Tyrfingsson T, Kiemeney LA, Mayordomo JI, Lindholt JS, Pedersen JH, Franklin WA, Wolf H, Montgomery GW, Heath AC, Martin NG, Madden PA, Giegling I, Rujescu D, Jarvelin MR, Salomaa V, Stumvoll M, Spector TD, Wichmann HE, Metspalu A, Samani NJ, Penninx BW, Oostra BA, Boomsma DI, Tiemeier H, van Duijn CM, Kaprio J, Gulcher JR, McCarthy MI, Peltonen L, Thorsteinsdottir U, Stefansson K (2010) Sequence variants at CHRNB3-CHRNA6 and CYP2A6 affect smoking behavior. Nat Genet 42(5):448–453

    Article  PubMed Central  PubMed  Google Scholar 

  • Town T, Abdullah L, Crawford F, Schinka J, Ordorica PI, Francis E, Hughes P, Duara R, Mullan M (1999) Association of a functional mu-opioid receptor allele (+118A) with alcohol dependency. Am J Med Genet 88(5):458–461

    Article  PubMed  Google Scholar 

  • Treutlein J, Cichon S, Ridinger M, Wodarz N, Soyka M, Zill P, Maier W, Moessner R, Gaebel W, Dahmen N, Fehr C, Scherbaum N, Steffens M, Ludwig KU, Frank J, Wichmann HE, Schreiber S, Dragano N, Sommer WH, Leonardi-Essmann F, Lourdusamy A, Gebicke-Haerter P, Wienker TF, Sullivan PF, Nothen MM, Kiefer F, Spanagel R, Mann K, Rietschel M (2009) Genome-wide association study of alcohol dependence. Arch Gen Psychiatry 66(7):773–784

    Article  PubMed Central  PubMed  Google Scholar 

  • Tsuang MT, Lyons MJ, Meyer JM, Doyle T, Eisen SA, Goldberg J, True W, Lin N, Toomey R, Eaves L (1998) Co-occurrence of abuse of different drugs in men: the role of drug-specific and shared vulnerabilities. Arch Gen Psychiatry 55(11):967–972

    Article  PubMed  Google Scholar 

  • Van den Oord EJ, Rujescu D, Robles JR, Giegling I, Birrell C, Bukszar J, Murrelle L, Moller HJ, Middleton L, Muglia P (2006) Factor structure and external validity of the PANSS revisited. Schizophr Res 82(2–3):213–223

    Article  PubMed  Google Scholar 

  • Vanyukov MM, Tarter RE, Kirisci L, Kirillova GP, Maher BS, Clark DB (2003) Liability to substance use disorders: 1. Common mechanisms and manifestations. Neurosci Biobehav Rev 27(6):507–515

    Article  PubMed  Google Scholar 

  • Vanyukov MM, Tarter RE, Kirillova GP, Kirisci L, Reynolds MD, Kreek MJ, Conway KP, Maher BS, Iacono WG, Bierut L, Neale MC, Clark DB, Ridenour TA (2012) Common liability to addiction and “gateway hypothesis”: theoretical, empirical and evolutionary perspective. Drug Alcohol Depend 123 Suppl 1S3-17

  • Wang YJ, Huang P, Ung A, Blendy JA, Liu-Chen LY (2012) Reduced expression of the mu opioid receptor in some, but not all, brain regions in mice with OPRM1 A112G. Neuroscience 205178-184

  • Wang YJ, Huang P, Blendy JA, Liu-Chen LY (2014) Brain region- and sex-specific alterations in DAMGO-stimulated [(35) S]GTPgammaS binding in mice with Oprm1 A112G. Addict Biol 19(3):354–361

    Article  PubMed Central  PubMed  Google Scholar 

  • Weiss RB, Baker TB, Cannon DS, von Nierderhausern A, Dunn DM, Matsunami N, Singh NA, Baird L, Coon H, McMahon WM, Piper ME, Fiore MC, Scholand MB, Connett JE, Kanner RE, Gahring LC, Rogers SW, Hoidal JR, Leppert MF (2008) A candidate gene approach identifies the CHRNA5-A3-B4 region as a risk factor for age-dependent nicotine addiction. PLoS Genet 4(7):e1000125

    Article  PubMed Central  PubMed  Google Scholar 

  • Wetherill L, Agrawal A, Kapoor M, Bertelsen S, Bierut LJ, Brooks A, Dick D, Hesselbrock M, Hesselbrock V, Koller DL, Le N, Nurnberger JI Jr, Salvatore JE, Schuckit M, Tischfield JA, Wang JC, Xuei X, Edenberg HJ, Porjesz B, Bucholz K, Goate AM, Foroud T (2015) Association of substance dependence phenotypes in the COGA sample. Addict Biol 20(3):617–627

    Article  PubMed  Google Scholar 

  • Xuei X, Flury-Wetherill L, Bierut L, Dick D, Nurnberger J Jr, Foroud T, Edenberg HJ (2007) The opioid system in alcohol and drug dependence: family-based association study. Am J Med Genet B Neuropsychiatr Genet 144B(7):877–884

    Article  PubMed  Google Scholar 

  • Zhang Y, Wang D, Johnson AD, Papp AC, Sadee W (2005) Allelic expression imbalance of human mu opioid receptor (OPRM1) caused by variant A118G. J Biol Chem 280(38):32618–32624

    Article  PubMed  Google Scholar 

  • Zhang H, Luo X, Kranzler HR, Lappalainen J, Yang BZ, Krupitsky E, Zvartau E, Gelernter J (2006a) Association between two mu-opioid receptor gene (OPRM1) haplotype blocks and drug or alcohol dependence. Hum Mol Genet 15(6):807–819

    Article  PubMed Central  PubMed  Google Scholar 

  • Zhang L, Kendler KS, Chen X (2006b) The mu-opioid receptor gene and smoking initiation and nicotine dependence. Behav Brain Funct 228

Download references

Acknowledgments

For facilitating this collaboration to meta-analyze rs1799971, we thank Jonathan Pollock and the National Institute on Drug Abuse, which provided infrastructure support through conference calls and meetings. For this project we wish to acknowledge and thank the following people. For meta-analysis coordination at Washington University: Weimin Duan. For administrative support at Washington University: Sherri Fisher. We also thank Michael Bruchas, Washington University, for helpful discussions. For CADD: principal investigators John Hewitt, Institute for Behavioral Genetics, University of Colorado Boulder; Michael Stallings, Institute for Behavioral Genetics, University of Colorado Boulder; Christian Hopfer, University of Denver; Sandra Brown, University of California San Diego. For COGA: Principal Investigators B. Porjesz, V. Hesselbrock, H. Edenberg, L. Bierut; COGA includes ten different centers: University of Connecticut (V. Hesselbrock); Indiana University (H.J. Edenberg, J. Nurnberger Jr., T. Foroud); University of Iowa (S. Kuperman, J. Kramer); SUNY Downstate (B. Porjesz); Washington University in St. Louis (L. Bierut, A. Goate, J. Rice, K. Bucholz); University of California at San Diego (M. Schuckit); Rutgers University (J. Tischfield); Texas Biomedical Research Institute (L. Almasy), Howard University (R. Taylor) and Virginia Commonwealth University (D. Dick). Other COGA collaborators include: L. Bauer (University of Connecticut); D. Koller, S. O’Connor, L. Wetherill, X. Xuei (Indiana University); Grace Chan (University of Iowa); S. Kang, N. Manz, M. Rangaswamy (SUNY Downstate); J. Rohrbaugh, J-C Wang (Washington University in St. Louis); A. Brooks (Rutgers University); and F. Aliev (Virginia Commonwealth University). A. Parsian and M. Reilly are the NIAAA Staff Collaborators. We continue to be inspired by our memories of Henri Begleiter and Theodore Reich, founding PI and Co-PI of COGA, and also owe a debt of gratitude to other past organizers of COGA, including Ting-Kai Li, currently a consultant with COGA, P. Michael Conneally, Raymond Crowe, and Wendy Reich, for their critical contributions. The authors thank Kim Doheny and Elizabeth Pugh from CIDR and Justin Paschall from the NCBI dbGaP staff for valuable assistance with genotyping and quality control in developing the dataset available at dbGaP. For COGEND: COGEND is a collaborative research group and a part of the NIDA Genetics Consortium. Michael Brent, Alison Goate, Dorothy Hatsukami, Anthony Hinrichs, Heidi Kromrei, Tracey Richmond, Joe Henry Steinbach, Jerry Stitzel, Scott Saccone, Sharon Murphy; in memory of Theodore Reich, founding Principal Investigator of COGEND, we are indebted to his leadership in the establishment and nurturing of COGEND and acknowledge with great admiration his seminal scientific contributions to the field. For GADD: John Hewitt, Institute for Behavioral Genetics, University of Colorado Boulder; Michael Stallings, Institute for Behavioral Genetics, University of Colorado Boulder; Christian Hopfer, University of Denver; Sandra Brown, University of California San Diego. For Kreek: Matthew Randesi. For NAG: Yi-Ling Chou. For Yale-Penn: Genotyping services for a part of Yale-Penn were provided by the Center for Inherited Disease Research (CIDR) and Yale University (Center for Genome Analysis).

Funding

R01 DA026911 from The National Institute on Drug Abuse (NIDA) supported this project and the coordinating team at Washington University. BG/ROMA are supported by R01 DA018823 (Todorov) from NIDA. CADD/GADD/NYS are supported by R01 DA021905 (Brown, Wall), R01 AA017889 (Ehringer), P60 DA011015 (Hewitt), R01 DA012845 (Hewitt), T32 DA017637 (Hewitt), R01 DA021913 (Hopfer), K24 DA032555 (Hopfer), K01 AA019447 (Kamens), and R01 DA035804 (Wall, Hopfer, Stallings) from NIDA and NIAAA. CATS is supported by R01 DA017305 (Nelson) from NIDA. Dr. Degenhardt is supported by an Australian National Health and Medical Research Council (NHMRC) principal research fellowship (#1041472). The National Drug and Alcohol Research Centre at UNSW Australia is supported by funding from the Australian Government under the Substance Misuse Prevention and Service Improvements Grant Fund. CEDAR-SADS is supported by R01 DA019157 (Vanyukov) from NIDA. CEDAR is supported by P50 DA005605 (Tarter) from NIDA. SADS is supported by R01 DA011922 (Vanyukov) from NIDA. The following studies made up the Cinciripini study: CASSI is supported by R01 DA11822 (Cinciripini), PEERS EMA is supported by K07 CA92209 (Carter), PEERS NS is supported by R21 CA81649 (Cinciripini) and PEERS WS, PEERS NS and SCOPE are supported by P50 CA070907 (Cinciripini) from NIDA and NCI. COGA is supported by U10 AA008401 (Bierut, Nurnberger, Edenberg, Hesselbrock, Porjesz) from NIAAA and genotyping was supported by U01 HG004438 (Valle) from The Genes and Environment Initiative (GEI) and HHSN268200782096C from the National Institutes of Health (NIH). Contributions from COGEND are supported by R01 DA019963 (Bierut), R01 DA013423 (Bierut), P01 CA089392 (Bierut), K08 DA030398 (L.-S. Chen), K08 DA032680 (Hartz), R01 DA019963 (Bierut), R01 DA038076 (L.-S. Chen), R21 DA038241 (Culverhouse), R01 DA026911 (Saccone) and R01 DA036583 (Bierut) from NIDA and the National Cancer Institute (NCI). Funding for COGEND genotyping was provided by 1 X01 HG005274-01 and performed at Center for Inherited Disease Research (CIDR) which is funded through a federal contract from NIH to JHU (HHSN268200782096C). Finnish Health 2000 is supported by Finnish National Institute for Health and Welfare institutional funding. FSCD is supported by R01 DA013423 (Bierut) from NIDA. Gene Environment Association Studies (GENEVA) Coordinating Center assisted with genotype cleaning as well as general study coordination, for FSCD; GENEVA is supported by U01 HG004446. FTC/FT12 is supported by K02 AA018755 (Rose), R01 AA-09203 (Rose), R37 AA-12502 (Rose), 141054 (Kaprio) and 263278 (Kaprio) from the National Institute on Alcohol Abuse and Alcoholism (NIAAA) and Academy of Finland. FTC/NAG-FIN is funded by GRAND (Kaprio) from Pfizer Inc., 213506 (Kaprio) and 129680 (Kaprio) from Academy of Finland, Health-F4-2007-201413 (Kaprio) from European Union Seventh Framework Programme, ENGAGE project, DA12854 (Madden) from NIDA, Wellcome Trust Sanger Institute, Sigrid Juselius Foundation and Jenny & Antti Wihuri Foundation. GESGA is supported by 01EB0410 (Mann), 01GS0852 (Kiefer), and  BMBF 01ZX1311A (Kiefer, Rietschel) (e:Med program) from the German Federal Ministry of Education and Research (BMBF), FZK 01GS08152 (Nöthen, Rietschel) from National Genome Research Network (NGFN Plus). Drs. Cichon and Nöthen are supported by Alfried Krupp von Bohlen und Halbach-Stiftung. IAS is supported by R01 DA015789 (Philibert) from NIDA. Kreek is supported by P50 DA05130 (Kreek) from NIDA and The Adelson Medical Research Foundation (Kreek). MCTFR is supported by R01 DA005147 (Iacono), R01 DA036216 (Iacono) and R01 U01 DA024417 (Iacono) from NIDA, R01 AA009367 (McGue) and R01 AA0011886 (McGue) from NIAAA and R01 MH066140 (McGue) from the National Institute of Mental Health (NIMH). MGHD is supported by R01 DA012846 (Tsuang) from NIDA and grants from NARSAD: The Brain and Behavior Research Foundation (Tsuang, Glatt), the Sidney R. Baer, Jr. Foundation (Tsuang), and the Gerber Foundation (Glatt). OYSUP is supported by RC2 DA028793 (Andrews) from NIDA. OZALC-NAG is supported by R01 AA075356 (Heath), R01 AA07728 (Heath), R01 AA13220 (Martin), R01 AA13321 (Heath), R01 AA13322 (Heath), R01 AA11998 (Heath) and R01 AA17688 (Heath) from NIAAA, R01 DA12854 (Madden) and K08 DA019951 (Pergadia) from NIDA and grants from the Australian National Health and Medical Research Council. Patch II/PiP are supported by C53/A6281 (Aveyard) from Cancer Research UK. Marcus Munafò and Paul Aveyard are members of the United Kingdom Centre for Tobacco and Alcohol Studies, a UKCRC Public Health Research: Centre of Excellence. Funding from British Heart Foundation, Cancer Research UK, Economic and Social Research Council, Medical Research Council, and the National Institute for Health Research, under the auspices of the UK Clinical Research Collaboration, is gratefully acknowledged. SMOFAM is supported by R01 DA003706 (Hops), U01 DA020830 (Lerman) from NIDA and 7PT2000-2004 (Swan) from University of California Tobacco-Related Disease Research Program. Utah is supported by P01 HL72903 (Hoidal) from NIDA and NHLBI. VA-Twin is supported by DA019498 (X. Chen) from NIDA; support was also provided by R01 DA18673 (Neale) from NIDA. Yale-Penn is supported by RC2 DA028909 (Gelernter), R01 DA12690 (Gelernter), R01 DA12849 (Gelernter), R01 DA18432 (Kranzler) and K01 DA24758 (Yang) from NIDA, R01 AA017535 (Gelernter) and R01 AA11330 (Gelernter) from NIAAA and Dr. Yang is supported by a Young Investigator Award from NARSAD: the Brain and Behavior Research Fund. Dr. Schwantes-An is also supported by the Division of Intramural Research Program of the National Human Genome Research Institute, National Institutes of Health. Funding sources had no further role in the study design; in the collection, analysis and interpretation of data; in the writing; or in the decision to submit the paper for publication.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nancy L. Saccone.

Ethics declarations

Conflict of interest

Dr. Bierut is listed as an inventor on Issued U.S. Patent 8 080 371, “Markers for Addiction” covering the use of certain SNPs in determining the diagnosis, prognosis, and treatment of addiction, and served as a consultant for Pfizer in 2008. Dr. NL Saccone is the spouse of Dr. SF Saccone, who is also listed as an inventor on the above patent. Dr. Cinciripini served on the scientific advisory board of Pfizer, conducted educational talks sponsored by Pfizer on smoking cessation (2006–2008), and has received grant support from Pfizer. Dr. Degenhardt has no relevant disclosures for this specific project; however, for general pharmaceutical company disclosures, Dr. Degenhardt has received untied educational grants from Reckitt Benckiser to conduct post-marketing surveillance of the diversion and injection of opioid substitution therapy medications in Australia. Although these activities are unrelated to the current study, Dr. Kranzler has been a consultant or advisory board member for Alkermes, Lilly, Lundbeck, Otsuka and Pfizer; he is also a member of the American Society of Clinical Psychopharmacology’s Alcohol Clinical Trials Initiative, which is supported by Ethypharm, Lilly, Lundbeck, AbbVie, and Pfizer. Dr. Ridinger is member of the advisory board of Lundbeck referring to Nalmefene. Prof. Dr. N. Scherbaum received honoraria for several activities (advisory boards, lectures, manuscripts and educational material) by the factories Sanofi-Aventis, Reckitt-Benckiser, Lundbeck, and Janssen-Cilag. During the last three years he participated in clinical trials financed by the pharmaceutical industry. The remaining authors declare no conflict of interest.

Informed Consent

The procedures followed were in accordance with the ethical standards of the responsible committee on human experimentation (institutional and national) and with the Helsinki Declaration of 1975, as revised in 2000 and 2008. All participants provided informed consent.

Additional information

Edited by Irwin Waldman.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schwantes-An, TH., Zhang, J., Chen, LS. et al. Association of the OPRM1 Variant rs1799971 (A118G) with Non-Specific Liability to Substance Dependence in a Collaborative de novo Meta-Analysis of European-Ancestry Cohorts. Behav Genet 46, 151–169 (2016). https://doi.org/10.1007/s10519-015-9737-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10519-015-9737-3

Keywords

Navigation