601
Views
15
CrossRef citations to date
0
Altmetric
Review

OPRM1 A118G Polymorphisms and Its Role in Opioid Addiction: Implication on Severity and Treatment Approaches

, ORCID Icon & ORCID Icon
Pages 361-368 | Published online: 27 Nov 2019

References

  • Kosten TR, George TP. The neurobiology of opioid dependence: implications for treatment. Sci Pract Perspect. 2002. doi:10.1151/spp021113
  • Hulse GK, English DR, Milne E, Holman CDJ. The quantification of mortality resulting from the regular use of illicit opiates. Addiction. 1999;94:221–229. doi:10.1046/j.1360-0443.1999.9422216.x10396790
  • Nutt D, King LA, Saulsbury W, Blakemore C. Development of a rational scale to assess the harm of drugs of potential misuse. Lancet. 2007;369:1047–1053. doi:10.1016/S0140-6736(07)60464-417382831
  • Bauer IE, Soares JC, Nielsen DA. The role of opioidergic genes in the treatment outcome of drug addiction pharmacotherapy: a systematic review. Am J Addict. 2015;24(1):15–23. doi:10.1111/ajad.1217225823631
  • Mercadante S. Prospects and challenges in opioid analgesia for pain management. Curr Med Res Opin. 2011;27(9):1741–1743. doi:10.1185/03007995.2011.60205721770715
  • Agrawal A, Verweij KJH, Gillespie NA, et al. The genetics of addiction-a translational perspective. Transl Psychiatry. 2012;2(7):e140. doi:10.1038/tp.2012.5422806211
  • Hancock DB, Markunas CA, Bierut LJ, Johnson EO. Human genetics of addiction: new insights and future directions. Curr Psychiatry Rep. 2018;20(2):8. doi:10.1007/s11920-018-0873-329504045
  • Oertel BG, Kettner M, Scholich K, et al. A common human μ-opioid receptor genetic variant diminishes the receptor signaling efficacy in brain regions processing the sensory information of pain. J Biol Chem. 2009;284:6530–6535. doi:10.1074/jbc.M80703020019116204
  • Shi J, Hui L, Xu Y, Wang F, Huang W, Hu G. Sequence variations in the mu-opioid receptor gene (OPRM1) associated with human addiction to heroin. Hum Mutat. 2002;19:459–460. doi:10.1002/humu.9026
  • Zhang D, Shao C, Shao M, et al. Effect of μ-opioid receptor gene polymorphisms on heroin-induced subjective responses in a chinese population. Biol Psychiatry. 2007;61:1244–1251. doi:10.1016/j.biopsych.2006.07.01217157823
  • Bond C, LaForge KS, Tian M, 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. doi:10.1073/pnas.95.16.96089689128
  • Browne CA, Erickson RL, Blendy JA, Lucki I. Genetic variation in the behavioral effects of buprenorphine in female mice derived from a murine model of the OPRM1 A118G polymorphism. Neuropharmacology. 2017;117:401–407. doi:10.1016/j.neuropharm.2017.02.00528188737
  • Chamorro AJ, Marcos M, Mirón-Canelo JA, Pastor I, González-Sarmiento R, Laso FJ. Association of μ-opioid receptor (OPRM1) gene polymorphism with response to naltrexone in alcohol dependence: a systematic review and meta-analysis. Addict Biol. 2012;17:505–512. doi:10.1111/j.1369-1600.2012.00442.x22515274
  • Li Y, Kantelip J-P, Schieveen PG, Davani S. Interindividual variability of methadone response. Mol Diagn Ther. 2008;12(2):109–124. doi:10.1007/BF0325627618422375
  • Berrettini W. A brief review of the genetics and pharmacogenetics of opioid use disorders. Dialogues Clin Neurosci. 2017;19:229.29302220
  • Kharasch ED, Regina KJ, Blood J, Friedel C. Methadone pharmacogenetics: CYP2B6 polymorphisms determine plasma concentrations, clearance, and metabolism. Anesthesiology. 2015;123:1142–1153. doi:10.1097/ALN.000000000000086726389554
  • Wang SC, Tsou HH, Ho IK, Lin KM, Liu YL. Pharmacogenomics study in a Taiwanese methadone maintenance cohort. J Food Drug Anal. 2013;21:S62–S68. doi:10.1016/j.jfda.2013.09.03625278738
  • Crowley JJ, Oslin DW, Patkar AA, et al. A genetic association study of the mu opioid receptor and severe opioid dependence. Psychiatr Genet. 2003;13:169–173. doi:10.1097/01.ypg.0000071762.90004.4512960749
  • Crist RC, Berrettini WH. Pharmacogenetics of OPRM1. Pharmacol Biochem Behav. 2014;123:25–33. doi:10.1016/j.pbb.2013.10.01824201053
  • Koob GF, Le Moal M. Addiction and the brain antireward system. Annu Rev Psychol. 2008;59:29–53. doi:10.1146/annurev.psych.59.103006.09354818154498
  • Le Merrer J, Becker JAJ, Befort K, Kieffer BL. Reward processing by the opioid system in the brain. Physiol Rev. 2009;89:1379–1412. doi:10.1152/physrev.00005.200919789384
  • Crabbe JC. Genetic contributions to addiction. Annu Rev Psychol. 2002;53:435–462. doi:10.1146/annurev.psych.53.100901.13514211752492
  • Nestler EJ. Genes and addiction. Nat Genet. 2000;26:277–281. doi:10.1038/8157011062465
  • Mistry CJ, Bawor M, Desai D, Marsh DC, Samaan Z. Genetics of opioid dependence: a review of the genetic contribution to opioid dependence. Curr Psychiatry Rev. 2014;10:156–167. doi:10.2174/157340051066614032000092825242908
  • Beer B, Erb R, Pavlic M, et al. Association of polymorphisms in pharmacogenetic candidate genes (OPRD1, GAL, ABCB1, OPRM1) with opioid dependence in european population: a case-control study. PLoS One. 2013;8:e75359. doi:10.1371/journal.pone.007535924086514
  • Fonseca F, de la Torre R, Díaz L, et al. Contribution of cytochrome P450 and ABCB1 genetic variability on methadone pharmacokinetics, dose requirements, and response. PLoS One. 2011;6:e19527. doi:10.1371/journal.pone.001952721589866
  • Ikeda K, Ide S, Han W, Hayashida M, Uhl GR, Sora I. How individual sensitivity to opiates can be predicted by gene analyses. Trends Pharmacol Sci. 2005;26:311–317. doi:10.1016/j.tips.2005.04.00115925706
  • Goldman D, Oroszi G, Ducci F. The genetics of addictions: uncovering the genes. Nat Rev Genet. 2005;6:521–532. doi:10.1038/nrg163515995696
  • Bart G, Kreek MJ, Ott J, et al. Increased attributable risk related to a functional μ-opioid receptor gene polymorphism in association with alcohol dependence in Central Sweden. Neuropsychopharmacology. 2005;30:417–422. doi:10.1038/sj.npp.130059815525999
  • Loh El W, Fann CS, Chang YT, Chang CJ, Cheng AT. Endogenous opioid receptor genes and alcohol dependence among Taiwanese Han. Alcohol Clin Exp Res. 2004. doi:10.1097/01.alc.0000106303.41755.b8
  • LaForge KS, Yuferov V, Kreek MJ. Opioid receptor and peptide gene polymorphisms: potential implications for addictions. Eur J Pharmacol. 2000;410:249–268. doi:10.1016/S0014-2999(00)00819-011134674
  • Kreek MJ. Pharmacogenetics and human molecular genetics of opiate and cocaine addictions and their treatments. Pharmacol Rev. 2005;57:1–26. doi:10.1124/pr.57.1.115734726
  • Oertel BG, Doehring A, Roskam B, et al. Genetic-epigenetic interaction modulates μ-opioid receptor regulation. Hum Mol Genet. 2012;21:4751–4760. doi:10.1093/hmg/dds31422875838
  • Zhang H, Luo X, Kranzler HR, et al. Association between two mu-opioid receptor gene (OPRM1) haplotype blocks and drug or alcohol dependence. Hum Mol Genet. 2006;15:807–819. doi:10.1093/hmg/ddl02416476706
  • Oueslati B, Moula O, Ghachem R. The impact of OPRM1’s genetic polymorphisms on methadone maintenance treatment in opioid addicts: a systematic review. Pharmacogenomics. 2018;19:741–747. doi:10.2217/pgs-2018-001729785888
  • Tsuang MT, Lyons MJ, Meyer JM, 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. doi:10.1001/archpsyc.55.11.9679819064
  • 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. doi:10.1176/appi.ajp.160.4.68712668357
  • Mura E, Govoni S, Racchi M, et al. Consequences of the 118A>G polymorphism in the OPRMI gene: translation from bench to bedside? J Pain Res. 2013;6:331–353. doi:10.2147/JPR.S4204023658496
  • Mague SD, Blendy JA. OPRM1 SNP (A118G): involvement in disease development, treatment response, and animal models. Drug Alcohol Depend. 2010;108(3):172–182. doi:10.1016/j.drugalcdep.2009.12.01620074870
  • Ahmed M, Ul Haq I, Faisal M, Waseem D, Taqi MM. Implication of OPRM1 A118G polymorphism in opioids addicts in Pakistan: in vitro and in silico analysis. J Mol Neurosci. 2018;65(4):472–479. doi:10.1007/s12031-018-1123-130033503
  • Kreek MJ, Reed B, Butelman ER. Current status of opioid addiction treatment and related preclinical research. Sci Adv. 2019;5(10):eaax9140. doi:10.1126/sciadv.aax914031616793
  • Tan E-C, Tan C-H, Karupathivan U, Yap EPH. Mu opioid receptor gene polymorphisms and heroin dependence in Asian populations. Neuroreport. 2003;14(4):569–572. doi:10.1097/00001756-200303240-0000812657887
  • Szeto CY, Tang NL, Lee DT, Stadlin A. Association between mu opioid receptor gene polymorphisms and Chinese heroin addicts. Neuroreport. 2001;12(6):1103–1106. doi:10.1097/00001756-200105080-0001111338173
  • Franke P, Wang T, Nöthen MM, et al. Nonreplication of association between mu-opioid-receptor gene (OPRM1) A118G polymorphism and substance dependence. Am J Med Genet. 2001;105(1):114–119.11424981
  • Hancock DB, Levy JL, Gaddis NC, et al. Cis-exzpression quantitative trait loci mapping reveals replicable associations with heroin addiction in OPRM1. Biol Psychiatry. 2015;78(7):474–484. doi:10.1016/j.biopsych.2015.01.00325744370
  • Schwantes-An T-H, Zhang J, Chen L-S, 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. 2016;46(2):151–169. doi:10.1007/s10519-015-9737-326392368
  • Domino EF, Evans CL, Ni L, Guthrie SK, Koeppe RA, Zubieta JK. Tobacco smoking produces greater striatal dopamine release in G-allele carriers with mu opioid receptor A118G polymorphism. Prog Neuro-Psychopharmacol Biol Psychiatry. 2012;38(2):236–240. doi:10.1016/j.pnpbp.2012.04.003
  • Sharafshah A, Fazel H, Albonaim A, et al. Association of OPRD1 gene variants with opioid dependence in addicted male individuals undergoing methadone treatment in the North of Iran. J Psychoactive Drugs. 2017;49(3):242–251. doi:10.1080/02791072.2017.129030328632076
  • Jones JD, Luba RR, Vogelman JL, Comer SD. Searching for evidence of genetic mediation of opioid withdrawal by opioid receptor gene polymorphisms. Am J Addict. 2016;25(1):41–48. doi:10.1111/ajad.1231626692286
  • Crist RC, Clarke TK, Ang A, et al. An intronic variant in OPRD1 predicts treatment outcome for opioid dependence in African-Americans. Neuropsychopharmacology. 2013;38(10):2003–2010. doi:10.1038/npp.2013.9923612435
  • Smith AH, Jensen KP, Li J, et al. Genome-wide association study of therapeutic opioid dosing identifies a novel locus upstream of OPRM1. Mol Psychiatry. 2017;22(3):346–352. doi:10.1038/mp.2016.25728115739
  • Wand GS, McCaul M, Yang X, et al. The mu-opioid receptor gene polymorphism (A118G) alters HPA axis activation induced by opioid receptor blockade. Neuropsychopharmacology. 2002;26(1):106–114. doi:10.1016/S0893-133X(01)00294-911751037
  • Hernandez-Avila CA, Wand G, Luo X, Gelernter J, Kranzler HR. Association between the cortisol response to opioid blockade and the Asn40Asp polymorphism at the mu-opioid receptor locus (OPRM1). Am J Med Genet B Neuropsychiatr Genet. 2003;118B(1):60–65. doi:10.1002/ajmg.b.1005412627468
  • Lötsch J, Skarke C, Liefhold J, Geisslinger G. Genetic predictors of the clinical response to opioid analgesics: clinical utility and future perspectives. Clin Pharmacokinet. 2004;43(14):983–1013. doi:10.2165/00003088-200443140-0000315530129
  • Klepstad P, Rakvåg TT, Kaasa S, et al. The 118 A > G polymorphism in the human μ-opioid receptor gene may increase morphine requirements in patients with pain caused by malignant disease. Acta Anaesthesiol Scand. 2004;48(10):1232–1239. doi:10.1111/j.1399-6576.2004.00517.x15504181
  • Mccance-Katz EF. (R)-methadone versus racemic methadone: what is best for patient care? Addiction. 2011;106(4):687–688. doi:10.1111/j.1360-0443.2011.03374.x21371149
  • Pérez de Los Cobos J, Siñol N, Trujols J, et al. Association of CYP2D6 ultrarapid metabolizer genotype with deficient patient satisfaction regarding methadone maintenance treatment. Drug Alcohol Depend. 2007;89:190–194. doi:10.1016/j.drugalcdep.2006.12.01817234366
  • Xie H, Griskevicius L, Ståhle L, et al. Pharmacogenetics of cyclophosphamide in patients with hematological malignancies. Eur J Pharm Sci. 2006;27:54–61. doi:10.1016/j.ejps.2005.08.00816183265
  • Lutfy K, Cowan A. Buprenorphine: a unique drug with complex pharmacology. Curr Neuropharmacol. 2004;2(4):395–402. doi:10.2174/157015904335947718997874
  • Mattick RP, Breen C, Kimber J, Davoli M. Buprenorphine maintenance versus placebo or methadone maintenance for opioid dependence. Cochrane Database Syst Rev. 2014;2014(2). doi:10.1002/14651858.CD002207.pub4
  • Soyka M. New developments in the management of opioid dependence: focus on sublingual buprenorphine–naloxone. Subst Abuse Rehabil. 2015;1. doi:10.2147/sar.s45585
  • Kirchmayer U, Davoli M, Verster AD, Amato L, Ferri A, Perucci CA. A systematic review on the efficacy of naltrexone maintenance treatment in opioid dependence. Addiction. 2002;97(10):1241–1249. doi:10.1046/j.1360-0443.2002.00217.x12359026
  • Krupitsky E, Nunes EV, Ling W, Gastfriend DR, Memisoglu A, Silverman BL. Injectable extended-release naltrexone (XR-NTX) for opioid dependence: long-term safety and effectiveness. Addiction. 2013;108(9):1628–1637. doi:10.1111/add.1220823701526
  • Mouly S, Bloch V, Peoc’H K, et al. Methadone dose in heroin-dependent patients: role of clinical factors, comedications, genetic polymorphisms and enzyme activity. Br J Clin Pharmacol. 2015;79:967–977. doi:10.1111/bcp.1257625556837
  • Ahmad T, Valentovic MA, Rankin GO. Effects of cytochrome P450 single nucleotide polymorphisms on methadone metabolism and pharmacodynamics. Biochem Pharmacol. 2018;153:196–204. doi:10.1016/j.bcp.2018.02.02029458047
  • Crettol S, Déglon JJ, Besson J, et al. ABCB1 and cytochrome P450 genotypes and phenotypes: influence on methadone plasma levels and response to treatment. Clin Pharmacol Ther. 2006. doi:10.1016/j.clpt.2006.09.012
  • Hustert E, Haberl M, Burk O, et al. The genetic determinants of the CYP3A5 polymorphism. Pharmacogenetics. 2001;11:773–779. doi:10.1097/00008571-200112000-0000511740341
  • Kuehl P, Zhang J, Lin Y, et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet. 2001;27:383–391. doi:10.1038/8688211279519
  • Lang T, Klein K, Fischer J, et al. Extensive genetic polymorphism in the human CYP2B6 gene with impact on expression and function in human liver. Pharmacogenetics. 2001;11:399–415. doi:10.1097/00008571-200107000-0000411470993
  • Pan YX. Alternative Pre-mRNA splicing of Mu opioid receptor gene: molecular mechanisms underlying the complex actions of Mu opioids. Pain Genet. 2013. doi:10.1002/9781118398890.ch6
  • Xu J, Lu Z, Narayan A, et al. Alternatively spliced mu opioid receptor C termini impact the diverse actions of morphine. J Clin Invest. 2017;127:1561–1573. doi:10.1172/JCI8876028319053
  • Gretton SK, Droney J. Splice variation of the mu-opioid receptor and its effect on the action of opioids. Br J Pain. 2014;8(4):133–138. doi:10.1177/204946371454711526516547
  • Hurd YL, O’Brien CP. Molecular genetics and new medication strategies for opioid addiction. Am J Psychiatry. 2018;175(10):935–942. doi:10.1176/appi.ajp.2018.1803035230068261
  • Soergel DG, Subach RA, Burnham N, et al. Biased agonism of the μ-opioid receptor by TRV130 increases analgesia and reduces on-target adverse effects versus morphine: a randomized, double-blind, placebo-controlled, crossover study in healthy volunteers. Pain. 2014;155(9):1829–1835. doi:10.1016/j.pain.2014.06.01124954166
  • Pan Y-X. Diversity and complexity of the Mu Opioid receptor gene: alternative Pre-mRNA splicing and promoters. DNA Cell Biol. 2005;24:736–750. doi:10.1089/dna.2005.24.73616274294
  • Xu J, Lu Z, Xu M, et al. A heroin addiction severity-associated intronic single nucleotide polymorphism modulates alternative Pre-mRNA splicing of the opioid receptor gene OPRM1 via hnRNPH interactions. J Neurosci. 2014;34:11048–11066. doi:10.1523/jneurosci.3986-13.201425122903
  • Vaillancourt K, Ernst C, Mash D, Turecki G. DNA methylation dynamics and cocaine in the brain: progress and prospects. Genes (Basel). 2017. doi:10.3390/genes8050138
  • Viet CT, Dang D, Aouizerat BE, et al. OPRM1 methylation contributes to opioid tolerance in cancer patients. J Pain. 2017;18:1046–1059. doi:10.1016/j.jpain.2017.04.00128456745
  • Ebrahimi G, Asadikaram G, Akbari H, et al. Elevated levels of DNA methylation at the OPRM1 promoter region in men with opioid use disorder. Am J Drug Alcohol Abuse. 2018. doi:10.1080/00952990.2016.1275659
  • Ferrer-Alcón M, La Harpe R, García-Sevilla JA. Decreased immunodensities of μ-opioid receptors, receptor kinases GRK 2/6 and β-arrestin-2 in postmortem brains of opiate addicts. Mol Brain Res. 2004;121:114–122. doi:10.1016/j.molbrainres.2003.11.00914969742
  • Bobadilla AC, Heinsbroek JA, Gipson CD, et al. Corticostriatal plasticity, neuronal ensembles, and regulation of drug-seeking behavior. Prog Brain Res. 2017. doi:10.1016/bs.pbr.2017.07.013
  • Kalivas PW. The glutamate homeostasis hypothesis of addiction. Nat Rev Neurosci. 2009;10:561–572. doi:10.1038/nrn251519571793