Generic placeholder image

Current Psychopharmacology

Editor-in-Chief

ISSN (Print): 2211-5560
ISSN (Online): 2211-5579

Review Article

In Search of Reward Deficiency Syndrome (RDS)-Free Controls: The “Holy Grail” in Genetic Addiction Risk Testing

Author(s): Kenneth Blum*, David Baron, Lisa Lott, Jessica V. Ponce, David Siwicki, Brent Boyett, Bruce Steinberg, Edward J. Modestino, Lyle Fried, Mary Hauser, Thomas Simpatico, Bill W. Downs, Thomas McLaughlin, Raju Hajela and Rajendra D. Badgaiyan

Volume 9, Issue 1, 2020

Page: [7 - 21] Pages: 15

DOI: 10.2174/2211556008666191111103152

Abstract

Background: The search for an accurate, gene-based test to identify heritable risk factors for Reward Deficiency Syndrome (RDS) was conducted based on hundreds of published studies about the role of dopamine in addictive behaviors, including risk for drug dependence and compulsive/impulsive behavior disorders. The term RDS was first coined by Blum’s group in 1995 to identify a group of behaviors with a common neurobiological mechanism associated with a polymorphic allelic propensity for hypodopaminergia.

Objectives: To outline the process used to select risk alleles of reward genes for the Genetic Addiction Risk Score (GARS) test. Consequently, to address the limitations caused by inconsistent results that occur in many case-control behavioral association studies. These limitations are perhaps due to the failure of investigators to adequately screen controls for drug and alcohol use disorder, and any of the many RDS behaviors, including nicotine dependence, obesity, pathological gambling, and internet gaming addiction.

Methods: Review of the literature related to the function of risk alleles of reward genes associated with hypodopaminergia relevant case-control association studies for the selection of alleles to be measured by the Genetic Addiction Risk Score (GARS) test.

Results: The prevalence of the DRD2 A1 allele in unscreened controls (33.3%), compared to “Super-Controls” [highly screened RDS controls (3.3%) in proband and family] is used to exemplify a possible solution.

Conclusion: Unlike One Gene-One Disease (OGOD), RDS is polygenetic, and very complex. In addition, any RDS-related behaviors must be eliminated from the control group in order to obtain the best possible statistical analysis instead of comparing the phenotype with diseaseridden controls.

Keywords: Behavioral genetic research, case controlled studies, genetic addiction association studies, genetic prevalence, hypodopaminergia, Reward Deficiency Syndrome (RDS), Single Nucleotide Polymorphisms (SNPs), study controls, super controls.

Graphical Abstract
[1]
Blum K, Chen ALC, Thanos PK, et al. Genetic addiction risk score (GARS) ™, a predictor of vulnerability to opioid dependence. Front Biosci (Elite Ed) 2018; 10: 175-96.
[http://dx.doi.org/10.2741/e816] [PMID: 28930612]
[2]
Blum K, Sheridan PJ, Wood RC, et al. The D2 dopamine receptor gene as a determinant of reward deficiency syndrome. J R Soc Med 1996; 89(7): 396-400.
[http://dx.doi.org/10.1177/014107689608900711] [PMID: 8774539]
[3]
Blum K, Kozlowski G. Ethanol and neuromodulator interactions: a cascade model of reward. Alcohol Behav 1990; pp. 131-50.
[4]
Erickson C. The Science of Addiction. New York: W.W. Norton 2007.
[5]
Blum K, Thanos PK, Badgaiyan RD, et al. Neurogenetics and gene therapy for reward deficiency syndrome: are we going to the Promised Land? Expert Opin Biol Ther 2015; 15(7): 973-85.
[http://dx.doi.org/10.1517/14712598.2015.1045871] [PMID: 25974314]
[6]
Li CY, Zhou WZ, Zhang PW, Johnson C, Wei L, Uhl GR. Meta-analysis and genome-wide interpretation of genetic susceptibility to drug addiction. BMC Genomics 2011; 12: 508.
[http://dx.doi.org/10.1186/1471-2164-12-508] [PMID: 21999673]
[7]
Koob GF, Volkow ND. Neurobiology of addiction: a neurocircuitry analysis. Lancet Psychiatry 2016; 3(8): 760-73.
[http://dx.doi.org/10.1016/S2215-0366(16)00104-8] [PMID: 27475769]
[8]
Noble EP, Blum K, Ritchie T, Montgomery A, Sheridan PJ. Allelic association of the D2 dopamine receptor gene with receptor-binding characteristics in alcoholism. Arch Gen Psychiatry 1991; 48(7): 648-54.
[http://dx.doi.org/10.1001/archpsyc.1991.01810310066012] [PMID: 2069496]
[9]
Kranzler HR, Smith RV, Schnoll R, Moustafa A, Greenstreet-Akman E. Precision medicine and pharmacogenetics: what does oncology have that addiction medicine does not? Addiction 2017; 112(12): 2086-94.
[http://dx.doi.org/10.1111/add.13818] [PMID: 28431457]
[10]
Gerra G, Somaini L, Leonardi C, et al. Association between gene variants and response to buprenorphine maintenance treatment. Psychiatry Res 2014; 215(1): 202-7.
[http://dx.doi.org/10.1016/j.psychres.2013.11.001] [PMID: 24274990]
[11]
Madrid GA, MacMurray J, Lee JW, Anderson BA, Comings DE. Stress as a mediating factor in the association between the DRD2 TaqI polymorphism and alcoholism. Alcohol 2001; 23(2): 117-22.
[http://dx.doi.org/10.1016/S0741-8329(00)00138-5] [PMID: 11331109]
[12]
Thompson MD, Kenna GA. Variation in the serotonin transporter gene and alcoholism: risk and response to pharmacotherapy. Alcohol Alcohol 2016; 51(2): 164-71.
[http://dx.doi.org/10.1093/alcalc/agv090] [PMID: 26311211]
[13]
Gold MS, Badgaiyan RD, Blum K. A Shared molecular and genetic basis for food and drug addiction: overcoming hypodopaminergic trait/state by incorporating dopamine agonistic therapy in psychiatry. Psychiatr Clin North Am 2015; 38(3): 419-62.
[http://dx.doi.org/10.1016/j.psc.2015.05.011] [PMID: 26300032]
[14]
Gold MS, Blum K, Oscar-Berman M, Braverman ER. Low dopamine function in attention deficit/hyperactivity disorder: should genotyping signify early diagnosis in children? Postgrad Med 2014; 126(1): 153-77.
[http://dx.doi.org/10.3810/pgm.2014.01.2735] [PMID: 24393762]
[15]
Batel P, Houchi H, Daoust M, Ramoz N, Naassila M, Gorwood P. A haplotype of the DRD1 gene is associated with alcohol dependence. Alcohol Clin Exp Res 2008; 32(4): 567-72.
[http://dx.doi.org/10.1111/j.1530-0277.2008.00618.x] [PMID: 18341651]
[16]
Kuo SC, Yeh YW, Chen CY, et al. DRD3 variation associates with early-onset heroin dependence, but not specific personality traits. Prog Neuropsychopharmacol Biol Psychiatry 2014; 51: 1-8.
[http://dx.doi.org/10.1016/j.pnpbp.2013.12.018] [PMID: 24398431]
[17]
Van Tol HH. Structural and functional characteristics of the dopamine D4 receptor. Adv Pharmacol 1998; 42: 486-90.
[http://dx.doi.org/10.1016/S1054-3589(08)60794-2] [PMID: 9327945]
[18]
Baransel Isir AB, Oguzkan S, Nacak M, Gorucu S, Dulger HE, Arslan A. The catechol-O-methyl transferase Val158Met polymorphism and susceptibility to cannabis dependence. Am J Forensic Med Pathol 2008; 29(4): 320-2.
[http://dx.doi.org/10.1097/PAF.0b013e3181847e56] [PMID: 19259017]
[19]
Wichers M, Aguilera M, Kenis G, et al. The catechol-O-methyl transferase Val158Met polymorphism and experience of reward in the flow of daily life. Neuropsychopharmacology 2008; 33(13): 3030-6.
[http://dx.doi.org/10.1038/sj.npp.1301520]
[20]
Ray R, Ruparel K, Newberg A, et al. Human Mu Opioid Receptor (OPRM1 A118G) polymorphism is associated with brain mu-opioid receptor binding potential in smokers. Proc Natl Acad Sci USA 2011; 108(22): 9268-73.
[http://dx.doi.org/10.1073/pnas.1018699108] [PMID: 21576462]
[21]
Byerley W, Hoff M, Holik J, Caron MG, Giros B. VNTR polymorphism for the human dopamine transporter gene (DAT1). Hum Mol Genet 1993; 2(3): 335.
[http://dx.doi.org/10.1093/hmg/2.3.335] [PMID: 8098980]
[22]
Contini V, Marques FZ, Garcia CE, Hutz MH, Bau CH. MAOA-uVNTR polymorphism in a Brazilian sample: further support for the association with impulsive behaviors and alcohol dependence. Am J Med Genet B Neuropsychiatr Genet 2006; 141B(3): 305-8.
[23]
Merenäkk L, Mäestu J, Nordquist N, et al. Effects of the serotonin transporter (5-HTTLPR) and α2A-adrenoceptor (C-1291G) genotypes on substance use in children and adolescents: a longitudinal study. Psychopharmacology (Berl) 2011; 215(1): 13-22.
[http://dx.doi.org/10.1007/s00213-010-2109-z] [PMID: 21140256]
[24]
van der Zwaluw CS, Engels RC, Vermulst AA, et al. A serotonin transporter polymorphism (5-HTTLPR) predicts the development of adolescent alcohol use. Drug Alcohol Depend 2010; 112(1-2): 134-9.
[http://dx.doi.org/10.1016/j.drugalcdep.2010.06.001] [PMID: 20598814]
[25]
Namkoong K, Cheon KA, Kim JW, Jun JY, Lee JY. Association study of dopamine D2, D4 receptor gene, GABAA receptor beta subunit gene, serotonin transporter gene polymorphism with children of alcoholics in Korea: a preliminary study. Alcohol 2008; 42(2): 77-81.
[http://dx.doi.org/10.1016/j.alcohol.2008.01.004] [PMID: 18358985]
[26]
Pearson-Fuhrhop KM, Dunn EC, Mortero S, et al. Dopamine genetic risk score predicts depressive symptoms in healthy adults and adults with depression. PLoS One 2014; 9(5)e93772
[http://dx.doi.org/10.1371/journal.pone.0093772] [PMID: 24834916]
[27]
Kawada Y, Hattori M, Fukuda R, Arai H, Inoue R, Nanko S. No evidence of linkage or association between tyrosine hydroxylase gene and affective disorder. J Affect Disord 1995; 34(2): 89-94.
[http://dx.doi.org/10.1016/0165-0327(95)00004-7] [PMID: 7665810]
[28]
Gold MS, Blum K, Febo M, et al. Molecular role of dopamine in anhedonia linked to reward deficiency syndrome (RDS) and anti- reward systems. Front Biosci (Schol Ed) 2018; 10: 309-25.
[http://dx.doi.org/10.2741/s518] [PMID: 29293435]
[29]
Chen TJ, Blum K, Mathews D, et al. Are dopaminergic genes involved in a predisposition to pathological aggression? Hypothesizing the importance of “super normal controls” in psychiatricgenetic research of complex behavioral disorders. Med Hypotheses 2005; 65(4): 703-7.
[http://dx.doi.org/10.1016/j.mehy.2005.04.037] [PMID: 15964153]
[30]
Chen AL, Blum K, Chen TJ, et al. Correlation of the Taq1 dopamine D2 receptor gene and percent body fat in obese and screened control subjects: a preliminary report. Food Funct 2012; 3(1): 40-8.
[http://dx.doi.org/10.1039/C1FO10089K] [PMID: 22051885]
[31]
Blum K. Reward Deficiency Syndrome. In: Wenzel A, Ed. The Sage Encyclopedia of Abnormal Clinical Psychology. Thousand Oaks, California: SAGE publishers 2017; pp. 4200.
[32]
Bolos AM, Dean M, Lucas-Derse S, Ramsburg M, Brown GL, Goldman D. Population and pedigree studies reveal a lack of association between the dopamine D2 receptor gene and alcoholism. JAMA 1990; 264(24): 3156-60.
[http://dx.doi.org/10.1001/jama.1990.03450240058040] [PMID: 1979357]
[33]
Kranzler HR, Zhou H, Kember RL, et al. Genome-wide association study of alcohol consumption and use disorder in 274,424 individuals from multiple populations. Nat Commun 2019; 10(1): 1499.
[http://dx.doi.org/10.1038/s41467-019-09480-8] [PMID: 30940813]
[34]
Blum K, Noble EP, Sheridan PJ, et al. Allelic association of human dopamine D2 receptor gene in alcoholism. JAMA 1990; 263(15): 2055-60.
[http://dx.doi.org/10.1001/jama.1990.03440150063027] [PMID: 1969501]
[35]
Prasad P, Ambekar A, Vaswani M. Case-control association analysis of dopamine receptor polymorphisms in alcohol dependence: a pilot study in Indian males. BMC Res Notes 2013; 6: 418.
[http://dx.doi.org/10.1186/1756-0500-6-418] [PMID: 24135011]
[36]
Verde Z, Santiago C, Rodríguez González-Moro JM, et al. ‘Smoking genes’: a genetic association study. PLoS One 2011; 6(10)e26668
[37]
Pinto JAF, Freitas PHB, Nunes FDD, Granjeiro PA, Santos LLD, Machado RM. Prevalence of polymorphisms in the ANKK1, DRD2, DRD3 genes and metabolic syndrome in refractory schizophrenia. Rev Lat Am Enfermagem 2018; 26e2983
[38]
Ding YC, Chi HC, Grady DL, et al. Evidence of positive selection acting at the human dopamine receptor D4 gene locus. Proc Natl Acad Sci USA 2002; 99(1): 309-14.
[http://dx.doi.org/10.1073/pnas.012464099] [PMID: 11756666]
[39]
van Dyck CH, Malison RT, Jacobsen LK, et al. Increased dopamine transporter availability associated with the 9-repeat allele of the SLC6A3 gene. J Nucl Med 2005; 46(5): 745-51.
[http://dx.doi.org/10.1073/pnas.012464099] [PMID: 15872345]
[40]
Saravani R, Galavi HR, Lotfian Sargazi M. Catechol-O-Methyltransferase (COMT) Gene (Val158Met) and Brain-Derived Neurotropic Factor (BDNF) (Val66Met) Genes Polymorphism in Schizophrenia: A Case-Control Study. Iran J Psychiatry 2017; 12(4): 265-70.
[41]
Hwang IW, Lim MH, Kwon HJ, Jin HJ. Association of Monoamine Oxidase A (MAOA) gene uVNTR and rs6323 polymorphisms with attention deficit and hyperactivity disorder in Korean children. Medicina (Kaunas) 2018; 54(3)E32
[http://dx.doi.org/10.3390/medicina54030032] [PMID: 30344263]
[42]
Haberstick BC, Smolen A, Williams RB, et al. Population frequencies of the triallelic 5HTTLPR in six ethnicially diverse samples from North America, Southeast Asia, and Africa. Behav Genet 2015; 45(2): 255-61.
[43]
Malhotra S, Basu D, Khullar M, Ghosh A, Chugh N. Candidate genes for alcohol dependence: A genetic association study from India. Indian J Med Res 2016; 144(5): 689-96.
[http://dx.doi.org/10.4103/ijmr.IJMR_1018_14] [PMID: 28361821]
[44]
Nobile B, Ramoz N, Jaussent I, et al. Polymorphism A118G of opioid receptor mu 1 (OPRM1) is associated with emergence of suicidal ideation at antidepressant onset in a large naturalistic cohort of depressed outpatients. Sci Rep 2019; 9(1): 2569.
[http://dx.doi.org/10.1038/s41598-019-39622-3] [PMID: 30796320]
[45]
Gade R, Blake H, MacMurray J, et al. Relationship of the GABRB3 gene to adult ADHD and personality traits in Caucasian and African-American samples. Psychiatr Genet 1996; 6: 164-5.
[46]
Gerra MC, Manfredini M, Cortese E, et al. Genetic and environmental risk factors for Cannabis use: preliminary results for the role of parental care perception. Subst Use Misuse 2019; 54(4): 670-80.
[http://dx.doi.org/10.1097/00041444-199623000-00081]
[47]
Shields PG, Lerman C, Audrain J, et al. Dopamine D4 receptors and the risk of cigarette smoking in African-Americans and Caucasians. Cancer Epidemiol Biomarkers Prev 1998; 7(6): 453-8.
[PMID: 9641486]
[48]
Li CY, Mao X, Wei L. Genes and (common) pathways underlying drug addiction. PLOS Comput Biol 2008; 4(1)e2
[http://dx.doi.org/10.1371/journal.pcbi.0040002] [PMID: 18179280]

© 2024 Bentham Science Publishers | Privacy Policy