The genetics of addiction-a translational perspective

A Agrawal, K J H Verweij, N A Gillespie, A C Heath, C N Lessov-Schlaggar, N G Martin, E C Nelson, W S Slutske, J B Whitfield, M T Lynskey, A Agrawal, K J H Verweij, N A Gillespie, A C Heath, C N Lessov-Schlaggar, N G Martin, E C Nelson, W S Slutske, J B Whitfield, M T Lynskey

Abstract

Addictions are serious and common psychiatric disorders, and are among the leading contributors to preventable death. This selective review outlines and highlights the need for a multi-method translational approach to genetic studies of these important conditions, including both licit (alcohol, nicotine) and illicit (cannabis, cocaine, opiates) drug addictions and the behavioral addiction of disordered gambling. First, we review existing knowledge from twin studies that indicates both the substantial heritability of substance-specific addictions and the genetic overlap across addiction to different substances. Next, we discuss the limited number of candidate genes which have shown consistent replication, and the implications of emerging genomewide association findings for the genetic architecture of addictions. Finally, we review the utility of extensions to existing methods such as novel phenotyping, including the use of endophenotypes, biomarkers and neuroimaging outcomes; emerging methods for identifying alternative sources of genetic variation and accompanying statistical methodologies to interpret them; the role of gene-environment interplay; and importantly, the potential role of genetic variation in suggesting new alternatives for treatment of addictions.

References

    1. American Psychiatric Association Diagnostic and Statistical Manual of Mental Disorders4th edn, Revised edn.American Psychiatric Association: Washington, DC; 1994
    1. Centers for Disease Control (CDC) Annual smoking-attributable mortality, years of potential life lost, and economic costs-United States. MMWR. 2002;51:300–303.
    1. Centers for Disease Control (CDC) Alcohol-Attributable Deaths and Years of Potential Life Lost—United States, 2001. MMWR. 2004;53:870.
    1. O'Brien C. Addiction and dependence in DSM-V. Addiction. 2011;106:866–867.
    1. Petry NM. Gambling and substance use disorders: current status and future directions. Am J Addict. 2007;16:1–9.
    1. Frascella J, Potenza MN, Brown LL, Childress AR. Shared brain vulnerabilities open the way for nonsubstance addictions: carving addiction at a new joint. Ann N Y Acad Sci. 2010;1187:294–315.
    1. Kendler KS, Neale MC, Sullivan P, Corey LA, Gardner CO, Prescott CA. A population-based twin study in women of smoking initiation and nicotine dependence. Psychol Med. 1999;29:299–308.
    1. Lessov CN, Martin NG, Statham DJ, Todorov AA, Slutske WS, Bucholz KK, et al. Defining nicotine dependence for genetic research: evidence from Australian twins. Psychol Med. 2004;34:865–879.
    1. Edwards AC, Maes HH, Pedersen NL, Kendler KS. A population-based twin study of the genetic and environmental relationship of major depression, regular tobacco use and nicotine dependence. Psychol Med. 2011;41:395–405.
    1. Lyons M, Hitsman B, Xian H, Panizzon MS, Jerskey BA, Santangelo S, et al. A twin study of smoking, nicotine dependence, and major depression in men. Nicotine Tob Res. 2008;10:97–108.
    1. Prescott CA, Kendler KS. Genetic and Environmental Influences on Alcohol and Tobacco Dependence among Women. NIAAA Res Monogr. 1995;30:59–87.
    1. Kendler KS, Heath AC, Neale MC, Kessler RC, Eaves LJ. A population-based twin study of alcoholism in women. JAMA. 1992;268:1877–1882.
    1. Heath AC, Martin NG. Genetic influences on alcohol consumption patterns and problem drinking: results from the Australian NH&MRC twin panel follow-up survey. Ann N Y Acad Sci. 1994;708:72–85.
    1. Heath AC, Whitfield JB, Madden PA, Bucholz KK, Dinwiddie SH, Slutske WS, et al. Towards a molecular epidemiology of alcohol dependence: analysing the interplay of genetic and environmental risk factors. Br J Psychiatry Suppl. 2001;40:s33–s40.
    1. Verweij KJ, Zietsch BP, Lynskey MT, Medland SE, Neale MC, Martin NG, et al. Genetic and environmental influences on cannabis use initiation and problematic use: a meta-analysis of twin studies. Addiction. 2010;105:417–430.
    1. Kendler KS, Karkowski LM, Neale MC, Prescott CA. Illicit psychoactive substance use, heavy use, abuse, and dependence in a US population-based sample of male twins. Arch Gen Psychiatry. 2000;57:261–269.
    1. Tsuang MT, Bar JL, Harley RM, Lyons MJ. The Harvard Twin Study of Substance Abuse: what we have learned. Harv Rev Psychiatry. 2001;9:267–279.
    1. van den Bree MB, Johnson EO, Neale MC, Pickens RW. Genetic and environmental influences on drug use and abuse/dependence in male and female twins. Drug Alcohol Depend. 1998;52:231–241.
    1. Tsuang MT, Lyons MJ, Meyer JM, Doyle T, Eisen SA, Goldberg J, et al. Co-occurrence of abuse of different drugs in men: the role of drug- specific and shared vulnerabilities. Arch Gen Psychiatry. 1998;55:967–972.
    1. Slutske WS, Zhu G, Meier MH, Martin NG. Genetic and environmental influences on disordered gambling in men and women. Arch Gen Psychiatry. 2010;67:624–630.
    1. Slutske WS, Eisen S, True WR, Lyons MJ, Goldberg J, Tsuang M. Common genetic vulnerability for pathological gambling and alcohol dependence in men. Arch Gen Psychiatry. 2000;57:666–673.
    1. Heath AC, Martin NG, Lynskey MT, Todorov AA, Madden PA. Estimating two-stage models for genetic influences on alcohol, tobacco or drug use initiation and dependence vulnerability in twin and family data. Twin Res. 2002;5:113–124.
    1. Neale MC, Harvey E, Maes HH, Sullivan PF, Kendler KS. Extensions to the modeling of initiation and progression: applications to substance use and abuse. Behav Genet. 2006;36:507–524.
    1. Agrawal A, Neale M, Jacobson K, Prescott CA, Kendler KS. Illicit drug use and abuse/dependence: modeling of two-stage variables using the CCC approach. Addict Behav. 2005;30:1043–1048.
    1. Heath AC, Lynskey MT, Waldron M.Substance useIn: Rutter M, Taylor E (eds).Handbook of Child and Adolescent Psychiatry Blackwell: London; 2007
    1. Dick DM, Pagan JL, Viken R, Purcell S, Kaprio J, Pulkkinen L, et al. Changing environmental influences on substance use across development. Twin Res Hum Genet. 2007;10:315–326.
    1. Kendler KS, Schmitt E, Aggen SH, Prescott CA. Genetic and environmental influences on alcohol, caffeine, cannabis, and nicotine use from early adolescence to middle adulthood. Arch Gen Psychiatry. 2008;65:674–682.
    1. Derringer J, Krueger RF, McGue M, Iacono WG. Genetic and environmental contributions to the diversity of substances used in adolescent twins: a longitudinal study of age and sex effects. Addiction. 2008;103:1744–1751.
    1. Derringer J, Krueger RF, Iacono WG, McGue M. Modeling the impact of age and sex on a dimension of poly-substance use in adolescence: a longitudinal study from 11- to 17-years-old. Drug Alcohol Depend. 2010;110:193–199.
    1. Rhee SH, Hewitt JK, Young SE, Corley RP, Crowley TJ, Stallings MC. Genetic and environmental influences on substance initiation, use, and problem use in adolescents. Arch Gen Psychiatry. 2003;60:1256–1264.
    1. Kendler KS, Jacobson KC, Prescott CA, Neale MC. Specificity of genetic and environmental risk factors for use and abuse/dependence of cannabis, cocaine, hallucinogens, sedatives, stimulants, and opiates in male twins. Am J Psychiatry. 2003;160:687–695.
    1. Kendler KS, Myers J, Prescott CA. Specificity of genetic and environmental risk factors for symptoms of cannabis, cocaine, alcohol, caffeine, and nicotine dependence. Arch Gen Psychiatry. 2007;64:1313–1320.
    1. Edenberg HJ. The genetics of alcohol metabolism: role of alcohol dehydrogenase and aldehyde dehydrogenase variants. Alcohol Res Health. 2007;30:5–13.
    1. Dick DM, Bierut LJ. The genetics of alcohol dependence. Curr Psychiatry Rep. 2006;8:151–157.
    1. McHugh RK, Hofmann SG, Asnaani A, Sawyer AT, Otto MW. The serotonin transporter gene and risk for alcohol dependence: a meta-analytic review. Drug Alcohol Depend. 2010;108:1–6.
    1. Edenberg HJ, Dick DM, Xuei X, Tian H, Almasy L, Bauer LO, et al. Variations in GABRA2, encoding the alpha 2 subunit of the GABA(A) receptor, are associated with alcohol dependence and with brain oscillations. Am J Hum Genet. 2004;74:705–714.
    1. Lappalainen J, Krupitsky E, Remizov M, Pchelina S, Taraskina A, Zvartau E, et al. Association between alcoholism and gamma-amino butyric acid alpha2 receptor subtype in a Russian population. Alcohol Clin Exp Res. 2005;29:493–498.
    1. Fehr C, Sander T, Tadic A, Lenzen KP, Anghelescu I, Klawe C, et al. Confirmation of association of the GABRA2 gene with alcohol dependence by subtype-specific analysis. Psychiatr Genet. 2006;16:9–17.
    1. Lind P, MacGregor S, Agrawal A, Heath AC, Martin NG, Whitfield JB. The role of GABRA2 in alcohol dependence, smoking and illicit drug use in an australian population sample. Alcohol Clin Exp Res. 2008;32:1721–1731.
    1. Covault J, Gelernter J, Hesselbrock V, Nellissery M, Kranzler HR. Allelic and haplotypic association of GABRA2 with alcohol dependence. Am J Med Genet B Neuropsychiatr Genet. 2004;129:104–109.
    1. Drgon T, D'Addario C, Uhl GR. Linkage disequilibrium, haplotype and association studies of a chromosome 4 GABA receptor gene cluster: candidate gene variants for addictions. Am J Med Genet B Neuropsychiatr Genet. 2006;141:854–860.
    1. Han S, Yang BZ, Kranzler HR, Oslin D, Anton R, Gelernter J. Association of CHRNA4 polymorphisms with smoking behavior in two populations. Am J Med Genet B Neuropsychiatr Genet. 2011;156B:421–429.
    1. Keskitalo-Vuokko K, Pitkaniemi J, Broms U, Heliovaara M, Aromaa A, Perola M, et al. Associations of nicotine intake measures with CHRN genes in finnish smokers. Nicotine Tob Res. 2011;13:686–690.
    1. Xie P, Kranzler HR, Krauthammer M, Cosgrove KP, Oslin D, Anton RF, et al. Rare nonsynonymous variants in Alpha-4 nicotinic acetylcholine receptor gene protect against nicotine dependence. Biol Psychiatry. 2011;70:528–536.
    1. Munafo MR, Timpson NJ, David SP, Ebrahim S, Lawlor DA. Association of the DRD2 gene Taq1A polymorphism and smoking behavior: a meta-analysis and new data. Nicotine Tob Res. 2009;11:64–76.
    1. Agrawal A, Lynskey MT. Candidate genes for cannabis use disorders: findings, challenges and directions. Addiction. 2009;104:518–532.
    1. Agrawal A, Edenberg HJ, Foroud T, Bierut LJ, Dunne G, Hinrichs AL, et al. Association of GABRA2 with drug dependence in the collaborative study of the genetics of alcoholism sample. Behav Genet. 2006;36:640–650.
    1. Corley RP, Zeiger JS, Crowley T, Ehringer MA, Hewitt JK, Hopfer CJ, et al. Association of candidate genes with antisocial drug dependence in adolescents. Drug Alcohol Depend. 2008;96:90–98.
    1. Gelernter J, Yu Y, Weiss R, Brady K, Panhuysen C, Yang BZ, et al. Haplotype spanning TTC12 and ANKK1, flanked by the DRD2 and NCAM1 loci, is strongly associated to nicotine dependence in two distinct American populations. Hum Mol Genet. 2006;15:3498–3507.
    1. Luo X, Kranzler H, Lappalainen J, Rosenheck R, Charney D, Zuo L, et al. CALCYON gene variation, schizophrenia, and cocaine dependence. Am J Med Genet B Neuropsychiatr Genet. 2004;125B:25–30.
    1. Kalayasiri R, Sughondhabirom A, Gueorguieva R, Coric V, Lynch WJ, Lappalainen J, et al. Dopamine beta-hydroxylase gene (DbetaH) -1021C → T influences self-reported paranoia during cocaine self-administration. Biol Psychiatry. 2007;61:1310–1313.
    1. Ittiwut R, Listman JB, Ittiwut C, Cubells JF, Weiss RD, Brady K, et al. Association between polymorphisms in catechol-O-methyltransferase (COMT) and cocaine-induced paranoia in European-American and African-American populations. Am J Med Genet B Neuropsychiatr Genet. 2011;156:651–660.
    1. Zhang H, Kranzler HR, Weiss RD, Luo X, Brady KT, Anton RF, et al. Pro-opiomelanocortin gene variation related to alcohol or drug dependence: evidence and replications across family- and population-based studies. Biol Psychiatry. 2009;66:128–136.
    1. Zuo L, Kranzler HR, Luo X, Yang BZ, Weiss R, Brady K, et al. Interaction between two independent CNR1 variants increases risk for cocaine dependence in European Americans: a replication study in family-based sample and population-based sample. Neuropsychopharmacology. 2009;34:1504–1513.
    1. Malison RT, Kranzler HR, Yang BZ, Gelernter J. Human clock, PER1 and PER2 polymorphisms: lack of association with cocaine dependence susceptibility and cocaine-induced paranoia. Psychiatr Genet. 2006;16:245–249.
    1. Dahl JP, Cubells JF, Ray R, Weller AE, Lohoff FW, Ferraro TN, et al. Analysis of variations in the tryptophan hydroxylase-2 (TPH2) gene in cocaine dependence. Addict Biol. 2006;11:76–83.
    1. Farrer LA, Kranzler HR, Yu Y, Weiss RD, Brady KT, Anton R, et al. Association of variants in MANEA with cocaine-related behaviors. Arch Gen Psychiatry. 2009;66:267–274.
    1. Grucza RA, Wang JC, Stitzel JA, Hinrichs AL, Saccone SF, Saccone NL, et al. A risk allele for nicotine dependence in CHRNA5 is a protective allele for cocaine dependence. Biol Psychiatry. 2008;64:922–929.
    1. Sherva R, Kranzler HR, Yu Y, Logue MW, Poling J, Arias AJ, et al. Variation in nicotinic acetylcholine receptor genes is associated with multiple substance dependence phenotypes. Neuropsychopharmacology. 2010;35:1921–1931.
    1. Uhl GR, Sora I, Wang Z. The mu opiate receptor as a candidate gene for pain: polymorphisms, variations in expression, nociception, and opiate responses. Proc Natl Acad Sci USA. 1999;96:7752–7755.
    1. Bond C, LaForge KS, Tian M, Melia D, Zhang S, Borg L, 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 U S A. 1998;95:9608–9613.
    1. Kroslak T, LaForge KS, Gianotti RJ, Ho A, Nielsen DA, Kreek MJ. The single nucleotide polymorphism A118G alters functional properties of the human mu opioid receptor. J Neurochem. 2007;103:77–87.
    1. LaForge KS, Yuferov V, Kreek MJ. Opioid receptor and peptide gene polymorphisms: potential implications for addictions. Eur J Pharmacol. 2000;410:249–268.
    1. Pan L, Xu J, Yu R, Xu MM, Pan YX, Pasternak GW. Identification and characterization of six new alternatively spliced variants of the human mu opioid receptor gene, Oprm. Neuroscience. 2005;133:209–220.
    1. Glatt SJ, Bousman C, Wang RS, Murthy KK, Rana BK, Lasky-Su JA, et al. Evaluation of OPRM1 variants in heroin dependence by family-based association testing and meta-analysis. Drug Alcohol Depend. 2007;90:159–165.
    1. Ray R, Doyle GA, Crowley JJ, Buono RJ, Oslin DW, Patkar AA, et al. A functional prodynorphin promoter polymorphism and opioid dependence. Psychiatr Genet. 2005;15:295–298.
    1. Comings DE, Blake H, Dietz G, Gade-Andavolu R, Legro RS, Saucier G, et al. The proenkephalin gene (PENK) and opioid dependence. Neuroreport. 1999;10:1133–1135.
    1. Gerra G, Leonardi C, Cortese E, D'Amore A, Lucchini A, Strepparola G, et al. Human kappa opioid receptor gene (OPRK1) polymorphism is associated with opiate addiction. Am J Med Genet B Neuropsychiatr Genet. 2007;144B:771–775.
    1. Zhang H, Kranzler HR, Yang BZ, Luo X, Gelernter J. The OPRD1 and OPRK1 loci in alcohol or drug dependence: OPRD1 variation modulates substance dependence risk. Mol Psychiatry. 2008;13:531–543.
    1. Franke P, Nothen MM, Wang T, Neidt H, Knapp M, Lichtermann D, et al. Human delta-opioid receptor gene and susceptibility to heroin and alcohol dependence. Am J Med Genet. 1999;88:462–464.
    1. Mayer P, Rochlitz H, Rauch E, Rommelspacher H, Hasse HE, Schmidt S, et al. Association between a delta opioid receptor gene polymorphism and heroin dependence in man. Neuroreport. 1997;8:2547–2550.
    1. Lobo DS, Kennedy JL. Genetic aspects of pathological gambling: a complex disorder with shared genetic vulnerabilities. Addiction. 2009;104:1454–1465.
    1. Comings DE, Gade R, Wu S, Chiu C, Dietz G, Muhleman D, et al. Studies of the potential role of the dopamine D1 receptor gene in addictive behaviors. Mol Psychiatry. 1997;2:44–56.
    1. da Silva Lobo DS, Vallada HP, Knight J, Martins SS, Tavares H, Gentil V, et al. Dopamine genes and pathological gambling in discordant sib-pairs. J Gambl Stud. 2007;23:421–433.
    1. Lobo DS, Souza RP, Tong RP, Casey DM, Hodgins DC, Smith GJ, et al. Association of functional variants in the dopamine D2-like receptors with risk for gambling behaviour in healthy Caucasian subjects. Biol Psychol. 2010;85:33–37.
    1. Thomasson HR, Crabb DW, Edenberg HJ, Li TK. Alcohol and aldehyde dehydrogenase polymorphisms and alcoholism. Behav Genet. 1993;23:131–136.
    1. Higuchi S, Matsushita S, Imazeki H, Kinoshita T, Takagi S, Kono H. Aldehyde dehydrogenase genotypes in Japanese alcoholics. Lancet. 1994;343:741–742.
    1. Lee HC, Lee HS, Jung SH, Yi SY, Jung HK, Yoon JH, et al. Association between polymorphisms of ethanol-metabolizing enzymes and susceptibility to alcoholic cirrhosis in a Korean male population. J Korean Med Sci. 2001;16:745–750.
    1. Koob GF, Volkow ND. Neurocircuitry of addiction. Neuropsychopharmacology. 2010;35:217–238.
    1. Volkow ND, Fowler JS, Wang GJ, Swanson JM, Telang F. Dopamine in drug abuse and addiction: results of imaging studies and treatment implications. Arch Neurol. 2007;64:1575–1579.
    1. Bierut LJ, Madden PA, Breslau N, Johnson EO, Hatsukami D, Pomerleau OF, et al. Novel genes identified in a high-density genome wide association study for nicotine dependence. Hum Mol Genet. 2007;16:24–35.
    1. Treutlein J, Cichon S, Ridinger M, Wodarz N, Soyka M, Zill P, et al. Genome-wide association study of alcohol dependence. Arch Gen Psychiatry. 2009;66:773–784.
    1. Frank J, Cichon S, Treutlein J, Ridinger M, Mattheisen M, Hoffmann P, et al. Genome-wide significant association between alcohol dependence and a variant in the ADH gene cluster. Addict Biol. 2012;17:171–180.
    1. Edenberg HJ, Koller DL, Xuei X, Wetherill L, McClintick JN, Almasy L, et al. Genome-wide association study of alcohol dependence implicates a region on chromosome 11. Alcohol Clin Exp Res. 2010;34:840–852.
    1. Bierut LJ, Agrawal A, Bucholz KK, Doheny KF, Laurie C, Pugh E, et al. A genome-wide association study of alcohol dependence. Proc Natl Acad Sci U S A. 2010;107:5082–5087.
    1. Heath AC, Whitfield JB, Martin NG, Pergadia ML, Goate AM, Lind PA, et al. A quantitative-trait genome-wide association study of alcoholism risk in the community: findings and implications. Biol Psychiatry. 2011;70:513–518.
    1. Agrawal A, Lynskey MT, Hinrichs A, Grucza R, Saccone SF, Krueger R, et al. A genome-wide association study of DSM-IV cannabis dependence. Addict Biol. 2011;16:514–518.
    1. Tobacco and Genetics Consortium Genome-wide meta-analyses identify multiple loci associated with smoking behavior. Nat Genet. 2010;42:441–447.
    1. Thorgeirsson TE, Gudbjartsson DF, Surakka I, Vink JM, Amin N, Geller F, et al. Sequence variants at CHRNB3-CHRNA6 and CYP2A6 affect smoking behavior. Nat Genet. 2010;42:448–453.
    1. Liu JZ, Tozzi F, Waterworth DM, Pillai SG, Muglia P, Middleton L, et al. Meta-analysis and imputation refines the association of 15q25 with smoking quantity. Nat Genet. 2010;42:436–440.
    1. Saccone SF, Hinrichs AL, Saccone NL, Chase GA, Konvicka K, Madden PA, et al. Cholinergic nicotinic receptor genes implicated in a nicotine dependence association study targeting 348 candidate genes with 3713 SNPs. Hum Mol Genet. 2007;16:36–49.
    1. Schumann G, Coin LJ, Lourdusamy A, Charoen P, Berger KH, Stacey D, et al. Genome-wide association and genetic functional studies identify autism susceptibility candidate 2 gene (AUTS2) in the regulation of alcohol consumption. Proc Natl Acad Sci U S A. 2011;108:7119–7124.
    1. Maher B. Personal genomes: the case of the missing heritability. Nature. 2008;456:18–21.
    1. Maher B.The search for the genome ‘Dark Matter' moves closer Nature News 20081235doi:10.1038/news.2008.1235
    1. Yang J, Benyamin B, McEvoy BP, Gordon S, Henders AK, Nyholt DR, et al. Common SNPs explain a large proportion of the heritability for human height. Nat Genet. 2010;42:565–569.
    1. Johnson C, Drgon T, Liu QR, Zhang PW, Walther D, Li CY, et al. Genome wide association for substance dependence: convergent results from epidemiologic and research volunteer samples. BMC Med Genet. 2008;9:113.
    1. Drgon T, Johnson CA, Nino M, Drgonova J, Walther DM, Uhl GR. ‘Replicated' genome wide association for dependence on illegal substances: genomic regions identified by overlapping clusters of nominally positive SNPs. Am J Med Genet B Neuropsychiatr Genet. 2011;156:125–138.
    1. Johnson C, Drgon T, Walther D, Uhl GR. Genomic regions identified by overlapping clusters of nominally-positive SNPs from genome-wide studies of alcohol and illegal substance dependence. PLoS One. 2011;6:e19210.
    1. Yang J, Manolio TA, Pasquale LR, Boerwinkle E, Caporaso N, Cunningham JM, et al. Genome partitioning of genetic variation for complex traits using common SNPs. Nat Genet. 2011;43:519–525.
    1. Davies G, Tenesa A, Payton A, Yang J, Harris SE, Liewald D, et al. Genome-wide association studies establish that human intelligence is highly heritable and polygenic. Mol Psychiatry. 2011;16:996–1005.
    1. Bierut LJ, Goate AM, Breslau N, Johnson EO, Bertelsen S, Fox L, et al. ADH1B is associated with alcohol dependence and alcohol consumption in populations of European and African ancestry. Mol Psychiatry. 2012;17:445–450.
    1. Agrawal A, Pergadia ML, Saccone SF, Lynskey MT, Wang JC, Martin NG, et al. An autosomal linkage scan for cannabis use disorders in the nicotine addiction genetics project. Arch Gen Psychiatry. 2008;65:713–721.
    1. Grant JD, Agrawal A, Bucholz KK, Madden PA, Pergadia ML, Nelson EC, et al. Alcohol consumption indices of genetic risk for alcohol dependence. Biol Psychiatry. 2009;66:795–800.
    1. Agrawal A, Grant JD, Littlefield A, Waldron M, Pergadia ML, Lynskey MT, et al. Developing a quantitative measure of alcohol consumption for genomic studies on prospective cohorts. J Stud Alcohol Drugs. 2009;70:157–168.
    1. Kendler KS, Myers J, Dick D, Prescott CA. The relationship between genetic influences on alcohol dependence and on patterns of alcohol consumption. Alcohol Clin Exp Res. 2010;34:1058–1065.
    1. Dick DM, Meyers JL, Rose RJ, Kaprio J, Kendler KS. Measures of current alcohol consumption and problems: two independent twin studies suggest a complex genetic architecture. Alcohol Clin Exp Res. 2011;35:2152–2161.
    1. Heatherton TF, Kozlowski LT, Frecker RC, Rickert W, Robinson J. Validity of the Fagerstrom test for nicotine dependence and of the Heaviness of Smoking Index among relatively light smokers. Br J Addict. 1989;84:791–799.
    1. Heatherton TF, Kozlowski LT, Frecker RC, Fagerstrom KO. The Fagerstrom test for nicotine dependence: a revision of the Fagerstrom Tolerance Questionnaire. Br J Addict. 1991;86:1119–1127.
    1. Moolchan ET, Aung AT, Henningfield JE. Treatment of adolescent tobacco smokers: issues and opportunities for exposure reduction approaches. Drug Alcohol Depend. 2003;70:223–232.
    1. Schuckit MA, Gold EO. A simultaneous evaluation of multiple markers of ethanol/placebo challenges in sons of alcoholics and controls. Arch Gen Psychiatry. 1988;45:211–216.
    1. Schuckit M.Vulnerability factors for alcoholismIn: Davis K (ed).Neuropsychopharmacology: The Fifth Generation of Progress Lippincott Williams and Wilkins: Baltimore; 2002
    1. Schuckit MA, Smith TL, Kalmijn J. The search for genes contributing to the low level of response to alcohol: patterns of findings across studies. Alcohol Clin Exp Res. 2004;28:1449–1458.
    1. Vogel-Sprott M, Chipperfield B. Family history of problem drinking among young male social drinkers: behavioral effects of alcohol. J Stud Alcohol. 1987;48:430–436.
    1. Ramchandani VA, Flury L, Morzorati SL, Kareken D, Blekher T, Foroud T, et al. Recent drinking history: association with family history of alcoholism and the acute response to alcohol during a 60 mg% clamp. J Stud Alcohol. 2002;63:734–744.
    1. Roh S, Matsushita S, Hara S, Maesato H, Matsui T, Suzuki G, et al. Role of GABRA2 in moderating subjective responses to alcohol. Alcohol Clin Exp Res. 2011;35:400–407.
    1. Pierucci-Lagha A, Covault J, Feinn R, Nellissery M, Hernandez-Avila C, Oncken C, et al. GABRA2 alleles moderate the subjective effects of alcohol, which are attenuated by finasteride. Neuropsychopharmacology. 2005;30:1193–1203.
    1. Hu X, Oroszi G, Chun J, Smith TL, Goldman D, Schuckit MA. An expanded evaluation of the relationship of four alleles to the level of response to alcohol and the alcoholism risk. Alcohol Clin Exp Res. 2005;29:8–16.
    1. Gottesman II, Gould TD. The endophenotype concept in psychiatry: etymology and strategic intentions. Am J Psychiatry. 2003;160:636–645.
    1. Kendler KS, Neale MC. Endophenotype: a conceptual analysis. Mol Psychiatry. 2010;15:789–797.
    1. Cannon TD, Keller MC. Endophenotypes in the genetic analyses of mental disorders. Annu Rev Clin Psychol. 2006;2:267–290.
    1. Field M, Cox WM. Attentional bias in addictive behaviors: a review of its development, causes, and consequences. Drug Alcohol Depend. 2008;97:1–20.
    1. Rangaswamy M, Porjesz B. Uncovering genes for cognitive (dys)function and predisposition for alcoholism spectrum disorders: a review of human brain oscillations as effective endophenotypes. Brain Res. 2008;1235:153–171.
    1. Porjesz B, Rangaswamy M. Neurophysiological endophenotypes, CNS disinhibition, and risk for alcohol dependence and related disorders. ScientificWorldJournal. 2007;7:131–141.
    1. Begleiter H, Porjesz B. Genetics of human brain oscillations. Int J Psychophysiol. 2006;60:162–171.
    1. Porjesz B, Almasy L, Edenberg HJ, Wang K, Chorlian DB, Foroud T, et al. Linkage disequilibrium between the beta frequency of the human EEG and a GABAA receptor gene locus. Proc Natl Acad Sci U S A. 2002;99:3729–3733.
    1. Kutalik Z, Benyamin B, Bergmann S, Mooser V, Waeber G, Montgomery GW, et al. Genome-wide association study identifies two loci strongly affecting transferrin glycosylation. Hum Mol Genet. 2011;20:3710–3717.
    1. Villafuerte S, Heitzeg MM, Foley S, Wendy Yau WY, Majczenko K, Zubieta JK, et al. Impulsiveness and insula activation during reward anticipation are associated with genetic variants in GABRA2 in a family sample enriched for alcoholism. Mol Psychiatry. 2012;17:511–519.
    1. Nikolova YS, Ferrell RE, Manuck SB, Hariri AR. Multilocus genetic profile for dopamine signaling predicts ventral striatum reactivity. Neuropsychopharmacology. 2011;36:1940–1947.
    1. Hamilton CM, Strader LC, Pratt JG, Maiese D, Hendershot T, Kwok RK, et al. The PhenX toolkit: get the most from your measures. Am J Epidemiol. 2011;174:253–260.
    1. Stankiewicz P, Lupski JR. Structural variation in the human genome and its role in disease. Annu Rev Med. 2010;61:437–455.
    1. 1000 Genomes Project Consortium A map of human genome variation from population-scale sequencing. Nature. 2010;467:1061–1073.
    1. Renthal W, Nestler EJ. Epigenetic mechanisms in drug addiction. Trends Mol Med. 2008;14:341–350.
    1. Maze I, Nestler EJ. The epigenetic landscape of addiction. Ann N Y Acad Sci. 2011;1216:99–113.
    1. Wong CC, Mill J, Fernandes C. Drugs and addiction: an introduction to epigenetics. Addiction. 2011;106:480–489.
    1. Bernstein BE, Stamatoyannopoulos JA, Costello JF, Ren B, Milosavljevic A, Meissner A, et al. The NIH roadmap epigenomics mapping consortium. Nat Biotechnol. 2010;28:1045–1048.
    1. Myers RM, Stamatoyannopoulos J, Snyder M, Dunham I, Hardison RC, Bernstein BE, et al. A user's guide to the encyclopedia of DNA elements (ENCODE) PLoS Biol. 2011;9:e1001046.
    1. Hurley JH, Ballard CJ, Edenberg HJ. Altering the relative abundance of GABA A receptor subunits changes GABA- and ethanol-responses in Xenopus oocytes. Alcohol Clin Exp Res. 2009;33:1089–1096.
    1. Liu J, Yang AR, Kelly T, Puche A, Esoga C, June HL, Jr, et al. Binge alcohol drinking is associated with GABAA alpha2-regulated Toll-like receptor 4 (TLR4) expression in the central amygdala. Proc Natl Acad Sci U S A. 2011;108:4465–4470.
    1. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, Bender D, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–575.
    1. Liu JZ, McRae AF, Nyholt DR, Medland SE, Wray NR, Brown KM, et al. A versatile gene-based test for genome-wide association studies. Am J Hum Genet. 2010;87:139–145.
    1. Raychaudhuri S, Plenge RM, Rossin EJ, Ng AC, Purcell SM, Sklar P, et al. Identifying relationships among genomic disease regions: predicting genes at pathogenic SNP associations and rare deletions. PLoS Genet. 2009;5:e1000534.
    1. Li MX, Gui HS, Kwan JS, Sham PC. GATES: a rapid and powerful gene-based association test using extended Simes procedure. Am J Hum Genet. 2011;88:283–293.
    1. Ruano D, Abecasis GR, Glaser B, Lips ES, Cornelisse LN, de Jong AP, et al. Functional gene group analysis reveals a role of synaptic heterotrimeric G proteins in cognitive ability. Am J Hum Genet. 2010;86:113–125.
    1. Reimers MA, Riley BP, Kalsi G, Kertes DA, Kendler KS.Pathway based analysis of genotypes in relation to alcohol dependence Pharmacogenomics J 2011(in press).
    1. Harris MA, Clark J, Ireland A, Lomax J, Ashburner M, Foulger R, et al. The Gene Ontology (GO) database and informatics resource. Nucleic Acids Res. 2004;32 (Database issue:D258–D261.
    1. Scarr S, McCartney J. How people make their own environment: a theory of genotype greater than environmental effects. Child Dev. 1983;54:424–435.
    1. Heath AC, Nelson EC. Effects of the interaction between genotype and environment. Research into the genetic epidemiology of alcohol dependence. Alcohol Res Health. 2002;26:193–201.
    1. Dick DM, Rose RJ, Viken RJ, Kaprio J, Koskenvuo M. Exploring gene-environment interactions: socioregional moderation of alcohol use. J Abnorm Psychol. 2001;110:625–632.
    1. Dick DM, Bernard M, Aliev F, Viken R, Pulkkinen L, Kaprio J, et al. The role of socioregional factors in moderating genetic influences on early adolescent behavior problems and alcohol use. Alcohol Clin Exp Res. 2009;33:1739–1748.
    1. Dick DM, Viken R, Purcell S, Kaprio J, Pulkkinen L, Rose RJ. Parental monitoring moderates the importance of genetic and environmental influences on adolescent smoking. J Abnorm Psychol. 2007;116:213–218.
    1. Button TM, Stallings MC, Rhee SH, Corley RP, Boardman JD, Hewitt JK. Perceived peer delinquency and the genetic predisposition for substance dependence vulnerability. Drug Alcohol Depend. 2009;100:1–8.
    1. Agrawal A, Balasubramanian S, Smith EK, Madden PA, Bucholz KK, Heath AC, et al. Peer substance involvement modifies genetic influences on regular substance involvement in young women. Addiction. 2010;105:1844–1853.
    1. Caspi A, Moffitt TE. Gene-environment interactions in psychiatry: joining forces with neuroscience. Nat Rev Neurosci. 2006;7:583–590.
    1. Rutter M, Moffitt TE, Caspi A. Gene-environment interplay and psychopathology: multiple varieties but real effects. J Child Psychol Psychiatry. 2006;47:226–261.
    1. Dick DM, Latendresse SJ, Lansford JE, Budde JP, Goate A, Dodge KA, et al. Role of GABRA2 in trajectories of externalizing behavior across development and evidence of moderation by parental monitoring. Arch Gen Psychiatry. 2009;66:649–657.
    1. Dick DM, Meyers JL, Latendresse SJ, Creemers HE, Lansford JE, Pettit GS, et al. CHRM2, parental monitoring, and adolescent externalizing behavior: evidence for gene-environment interaction. Psychol Sci. 2011;22:481–489.
    1. Nelson EC, Agrawal A, Pergadia ML, Wang JC, Whitfield JB, Saccone FS, et al. H2 haplotype at chromosome 17q21.31 protects against childhood sexual abuse-associated risk for alcohol consumption and dependence. Addict Biol. 2010;15:1–11.
    1. Nelson EC, Heath AC, Lynskey MT, Bucholz KK, Madden PA, Statham DJ, et al. Childhood sexual abuse and risks for licit and illicit drug-related outcomes: a twin study. Psychol Med. 2006;36:1473–1483.
    1. Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc Natl Acad Sci U S A. 2005;102:10604–10609.
    1. Risch N, Herrell R, Lehner T, Liang KY, Eaves L, Hoh J, et al. Interaction between the serotonin transporter gene (5-HTTLPR), stressful life events, and risk of depression: a meta-analysis. JAMA. 2009;301:2462–2471.
    1. Duncan LE, Keller MC. A critical review of the first 10 years of candidate gene-by-environment interaction research in psychiatry. Am J Psychiatry. 2011;168:1041–1049.
    1. van Ijzendoorn MH, Bakermans-Kranenburg MJ, Belsky J, Beach S, Brody G, Dodge KA, et al. Gene-by-environment experiments: a new approach to finding the missing heritability. Nat Rev Genet. 2011;12:881.
    1. Schnoll RA, Lerman C. Current and emerging pharmacotherapies for treating tobacco dependence. Expert Opin Emerg Drugs. 2006;11:429–444.
    1. Hughes JR. How confident should we be that smoking cessation treatments work. Addiction. 2009;104:1637–1640.
    1. Sturgess JE, George TP, Kennedy JL, Heinz A, Muller DJ. Pharmacogenetics of alcohol, nicotine and drug addiction treatments. Addict Biol. 2011;16:357–376.
    1. Oslin DW, Berrettini W, Kranzler HR, Pettinati H, Gelernter J, Volpicelli JR, et al. A functional polymorphism of the mu-opioid receptor gene is associated with naltrexone response in alcohol-dependent patients. Neuropsychopharmacology. 2003;28:1546–1552.
    1. Ray LA, Hutchison KE. Effects of naltrexone on alcohol sensitivity and genetic moderators of medication response: a double-blind placebo-controlled study. Arch Gen Psychiatry. 2007;64:1069–1077.
    1. Anton RF, Oroszi G, O'Malley S, Couper D, Swift R, Pettinati H, 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. 2008;65:135–144.
    1. Heilig M, Goldman D, Berrettini W, O'Brien CP. Pharmacogenetic approaches to the treatment of alcohol addiction. Nat Rev Neurosci. 2011;12:670–684.
    1. Kenna GA. Medications acting on the serotonergic system for the treatment of alcohol dependent patients. Curr Pharm Des. 2010;16:2126–2135.
    1. McKay JD, Truong T, Gaborieau V, Chabrier A, Chuang SC, Byrnes G, et al. A genome-wide association study of upper aerodigestive tract cancers conducted within the INHANCE consortium. PLoS Genet. 2011;7:e1001333.
    1. Brooks PJ, Enoch MA, Goldman D, Li TK, Yokoyama A. The alcohol flushing response: an unrecognized risk factor for esophageal cancer from alcohol consumption. PLoS Med. 2009;6:e50.
    1. Brooks PJ, Goldman D, Li TK. Alleles of alcohol and acetaldehyde metabolism genes modulate susceptibility to oesophageal cancer from alcohol consumption. Hum Genomics. 2009;3:103–105.
    1. Wang Y, Broderick P, Webb E, Wu X, Vijayakrishnan J, Matakidou A, et al. Common 5p15.33 and 6p21.33 variants influence lung cancer risk. Nat Genet. 2008;40:1407–1409.
    1. Thorgeirsson TE, Geller F, Sulem P, Rafnar T, Wiste A, Magnusson KP, et al. A variant associated with nicotine dependence, lung cancer and peripheral arterial disease. Nature. 2008;452:638–642.
    1. Hung RJ, McKay JD, Gaborieau V, Boffetta P, Hashibe M, Zaridze D, et al. A susceptibility locus for lung cancer maps to nicotinic acetylcholine receptor subunit genes on 15q25. Nature. 2008;452:633–637.
    1. Saccone NL, Culverhouse RC, Schwantes-An TH, Cannon DS, Chen X, Cichon S, et al. Multiple independent loci at chromosome 15q25.1 affect smoking quantity: a meta-analysis and comparison with lung cancer and COPD PLoS Genet 20106pii: e1001053.
    1. Wacholder S, Chatterjee N, Caporaso N. Intermediacy and gene-environment interaction: the example of CHRNA5-A3 region, smoking, nicotine dependence, and lung cancer. J Natl Cancer Inst. 2008;100:1488–1491.
    1. Calandrella D, Antonini A. Pathological gambling in Parkinson's disease: disease related or drug related. Expert Rev Neurother. 2011;11:809–814.
    1. Bell RL, Rodd ZA, Lumeng L, Murphy JM, McBride WJ. The alcohol-preferring P rat and animal models of excessive alcohol drinking. Addict Biol. 2006;11:270–288.
    1. Sommer W, Hyytia P, Kiianmaa K. The alcohol-preferring AA and alcohol-avoiding ANA rats: neurobiology of the regulation of alcohol drinking. Addict Biol. 2006;11:289–309.
    1. Crabbe JC, Harris RA, Koob GF. Preclinical studies of alcohol binge drinking. Ann N Y Acad Sci. 2011;1216:24–40.
    1. Heberlein U. Genetics of alcohol-induced behaviors in Drosophila. Alcohol Res Health. 2000;24:185–188.
    1. Crabbe JC. Consilience of rodent and human phenotypes relevant for alcohol dependence. Addict Biol. 2010;15:103–108.
    1. Merikangas KR, Risch N. Genomic priorities and public health. Science. 2003;302:599–601.
    1. Hirschhorn JN. Genomewide association studies—illuminating biologic pathways. N Engl J Med. 2009;360:1699–1701.
    1. Oze I, Matsuo K, Wakai K, Nagata C, Mizoue T, Tanaka K, et al. Alcohol drinking and esophageal cancer risk: an evaluation based on a systematic review of epidemiologic evidence among the Japanese population. Jpn J Clin Oncol. 2011;41:677–692.
    1. Luo X, Kranzler HR, Zuo L, Wang S, Schork NJ, Gelernter J. Diplotype trend regression analysis of the ADH gene cluster and the ALDH2 gene: multiple significant associations with alcohol dependence. Am J Hum Genet. 2006;78:973–987.
    1. Macgregor S, Lind PA, Bucholz KK, Hansell NK, Madden PA, Richter MM, et al. Associations of ADH and ALDH2 gene variation with self report alcohol reactions, consumption and dependence: an integrated analysis. Hum Mol Genet. 2009;18:580–593.
    1. Li D, Zhao H, Gelernter J. Strong Association of the Alcohol Dehydrogenase 1B Gene (ADH1B) with alcohol dependence and alcohol-induced medical diseases. Biol Psychiatry. 2011;70:504–512.
    1. Soyka M, Preuss UW, Hesselbrock V, Zill P, Koller G, Bondy B. GABA-A2 receptor subunit gene (GABRA2) polymorphisms and risk for alcohol dependence. J Psychiatr Res. 2008;42:184–191.
    1. Noble EP, Blum K. Alcoholism and the D2 dopamine receptor gene. JAMA. 1993;270:1547–1548.
    1. Munafo MR, Matheson IJ, Flint J. Association of the DRD2 gene Taq1A polymorphism and alcoholism: a meta-analysis of case-control studies and evidence of publication bias. Mol Psychiatry. 2007;12:454–461.
    1. Smith L, Watson M, Gates S, Ball D, Foxcroft D. Meta-analysis of the association of the Taq1A polymorphism with the risk of alcohol dependency: a HuGE gene-disease association review. Am J Epidemiol. 2008;167:125–138.
    1. Bierut LJ, Stitzel JA, Wang JC, Hinrichs AL, Grucza RA, Xuei X, et al. Variants in nicotinic receptors and risk for nicotine dependence. Am J Psychiatry. 2008;165:1163–1171.
    1. Kuryatov A, Berrettini W, Lindstrom J. Acetylcholine receptor (AChR) alpha5 subunit variant associated with risk for nicotine dependence and lung cancer reduces (alpha4beta2)alpha5 AChR function. Mol Pharmacol. 2011;79:119–125.
    1. Lessov-Schlaggar CN, Pergadia ML, Khroyan TV, Swan GE. Genetics of nicotine dependence and pharmacotherapy. Biochem Pharmacol. 2008;75:178–195.
    1. Heath AC. Persist or quit? Testing for a genetic contribution to smoking persistence. Acta Genet Med Gemellol (Roma) 1990;39:447–458.
    1. Hughes JR. Should criteria for drug dependence differ across drugs. Addiction. 2006;101 (Suppl 1:134–141.
    1. McBride O, Strong DR, Kahler CW. Exploring the role of a nicotine quantity-frequency use criterion in the classification of nicotine dependence and the stability of a nicotine dependence continuum over time. Nicotine Tob Res. 2010;12:207–216.
    1. Strong DR, Kahler CW, Colby SM, Griesler PC, Kandel D. Linking measures of adolescent nicotine dependence to a common latent continuum. Drug Alcohol Depend. 2009;99:296–308.
    1. Bierut LJ, Dinwiddie SH, Begleiter H, Crowe RR, Hesselbrock V, Nurnberger JI, Jr, et al. 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. 1998;55:982–988.
    1. Kaprio J, Hammar N, Koskenvuo M, Floderus-Myrhed B, Langinvainio H, Sarna S. Cigarette smoking and alcohol use in Finland and Sweden: a cross-national twin study. Int J Epidemiol. 1982;11:378–386.
    1. Pergadia ML, Heath AC, Martin NG, Madden PA. Genetic analysis of DSM-IV nicotine withdrawal in adult twins. Psychol Med. 2006;36:963–972.
    1. Han S, Gelernter J, Luo X, Yang BZ. Meta-analysis of 15 genome-wide linkage scans of smoking behavior. Biol Psychiatry. 2010;67:12–19.
    1. Boardman JD. State-level moderation of genetic tendencies to smoke. Am J Public Health. 2009;99:480–486.
    1. Johnson EO, Chen LS, Breslau N, Hatsukami D, Robbins T, Saccone NL, et al. Peer smoking and the nicotinic receptor genes: an examination of genetic and environmental risks for nicotine dependence. Addiction. 2010;105:2014–2022.
    1. Chen LS, Johnson EO, Breslau N, Hatsukami D, Saccone NL, Grucza RA, et al. Interplay of genetic risk factors and parent monitoring in risk for nicotine dependence. Addiction. 2009;104:1731–1740.
    1. Weiss RB, Baker TB, Cannon DS, von NA, Dunn DM, Matsunami N, et al. A candidate gene approach identifies the CHRNA5-A3-B4 region as a risk factor for age-dependent nicotine addiction. PLoS Genet. 2008;4:e1000125.
    1. Grucza RA, Johnson EO, Krueger RF, Breslau N, Saccone NL, Chen LS, et al. Incorporating age at onset of smoking into genetic models for nicotine dependence: evidence for interaction with multiple genes. Addict Biol. 2010;15:346–357.
    1. Vink JM, Smit AB, de Geus EJ, Sullivan P, Willemsen G, Hottenga JJ, et al. Genome-wide association study of smoking initiation and current smoking. Am J Hum Genet. 2009;84:367–379.
    1. Jackson KJ, Marks MJ, Vann RE, Chen X, Gamage TF, Warner JA, et al. Role of alpha5 nicotinic acetylcholine receptors in pharmacological and behavioral effects of nicotine in mice. J Pharmacol Exp Ther. 2010;334:137–146.
    1. Fowler CD, Lu Q, Johnson PM, Marks MJ, Kenny PJ. Habenular alpha5 nicotinic receptor subunit signalling controls nicotine intake. Nature. 2011;471:597–601.
    1. Hong LE, Hodgkinson CA, Yang Y, Sampath H, Ross TJ, Buchholz B, et al. A genetically modulated, intrinsic cingulate circuit supports human nicotine addiction. Proc Natl Acad Sci U S A. 2010;107:13509–13514.
    1. Swan GE, Javitz HS, Jack LM, Wessel J, Michel M, Hinds DA, et al. Varenicline for smoking cessation: nausea severity and variation in nicotinic receptor genes Pharmacogenomics J 2011(in press).
    1. Rose JE, Behm FM, Drgon T, Johnson C, Uhl GR. Personalized smoking cessation: interactions between nicotine dose, dependence and quit-success genotype score. Mol Med. 2010;16:247–253.

Source: PubMed

3
Prenumerera