1
|
Ide S, Iwase N, Arai K, Kojima M, Ushiyama S, Ikeda K. Inhibitory effects of the selective μ-opioid receptor antagonist UD-030 on methamphetamine-induced conditioned place preference. Neuropsychopharmacol Rep 2025; 45:e12503. [PMID: 39601072 DOI: 10.1002/npr2.12503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Although methamphetamine (METH) and other addictive substance use disorders are a major social problem worldwide, appropriate pharmacotherapies have not yet been discovered. Subtype-nonselective opioid receptor antagonists, such as naltrexone (NTX), have been reported to suppress METH addiction, but unclear are the opioid receptor subtypes that are involved in this beneficial effect. To clarify the role of μ-opioid receptors (MOPs), we examined effects of the novel nonpeptidic MOP-selective antagonist UD-030 on the acquisition and expression of METH-induced conditioned place preference (CPP) using behavioral tests in C57BL/6J mice. UD-030 was found to inhibit both the acquisition and expression of METH-induced CPP in a dose-dependent manner, with effects comparable to those observed with NTX. These findings suggest that UD-030 has the potential to mitigate METH-related reward mechanisms and may serve as a promising candidate for MOP-selective pharmacotherapy targeting METH addiction.
Collapse
Affiliation(s)
- Soichiro Ide
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | | | | | | | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Department of Neuropsychopharmacology, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
2
|
Hriatpuii V, Sema HP, Vankhuma C, Iyer M, Subramaniam MD, Rao KRSS, Vellingiri B, Kumar NS. Association of OPRM1 with addiction: a review on drug, alcohol and smoking addiction in worldwide population. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00249-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Drugs are chemicals which can disrupt the nerve cell functions of the brain. The present study aims to investigate the addiction related gene (OPRM1) in three types of addiction—drugs, alcohol and smoking. Pathway for the addiction was ascertained through KEGG database, and the hotspot mutations for various populations were identified from Gnomad-exomes database. In silico analyses like SIFT, Polyphen, Hope, I-mutant and mutation taster were performed to understand the amino acid substitution, protein function, stability and pathogenicity of the variants.
Main body
Addiction-related variants were found in exons 1, 2 and 3, while the exon 4 did not exhibit any addiction related variation. Among all the variants from this gene, rs1799971 (A118G) polymorphism was the most commonly studied variation for addiction in different populations worldwide. Population-wise allele and genotype frequencies, demographic and epidemiological studies have also been performed from different populations, and the possible association of these variants with addiction was evaluated.
Conclusion
Our findings suggest that OPRM1 polymorphism impact as pharmacogenetic predictor of response to naltrexone and can also address the genetic predisposition related to addiction in human beings.
Collapse
|
3
|
Guerin AA, Nestler EJ, Berk M, Lawrence AJ, Rossell SL, Kim JH. Genetics of methamphetamine use disorder: A systematic review and meta-analyses of gene association studies. Neurosci Biobehav Rev 2021; 120:48-74. [PMID: 33217458 PMCID: PMC7856253 DOI: 10.1016/j.neubiorev.2020.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 02/01/2023]
Abstract
Genetic susceptibility to methamphetamine use disorder is poorly understood. No twin or adequately powered genome-wide association studies (GWASs) have been conducted. However, there are a large number of hypothesis-driven candidate gene association studies, which were systematically reviewed herein. Seventy-six studies were identified, investigating markers of 75 different genes. Allele frequencies, odds ratios, 95 % confidence intervals and power were calculated. Risk of bias was also assessed as a quality measure. Meta-analyses were conducted for gene markers if three or more studies were available. Eleven markers from adequately powered studies were significantly associated with methamphetamine use disorder, with Fatty Acid Amide Hydrolase (FAAH) and Brain Derived Neurotrophic Factor (BDNF) representing promising targets. Limitations of these studies include unclear rationale for candidate gene selection, low power and high risk of bias. Future research should include replications to enable more meta-analyses, well-powered GWASs or whole exome or genome sequencing, as well as twin and family studies to further complement the findings of this review to uncover genetic contributions toward methamphetamine use disorder.
Collapse
Affiliation(s)
- Alexandre A Guerin
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Berk
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Andrew J Lawrence
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Susan L Rossell
- Centre for Mental Health, Swinburne University of Technology, Melbourne, VIC, Australia; Department of Psychiatry, St Vincent's Hospital, Melbourne, VIC, Australia
| | - Jee Hyun Kim
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, 3052, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3052, Australia; Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia.
| |
Collapse
|
4
|
Stafford AM, Reed C, Baba H, Walter NAR, Mootz JRK, Williams RW, Neve KA, Fedorov LM, Janowsky AJ, Phillips TJ. Taar1 gene variants have a causal role in methamphetamine intake and response and interact with Oprm1. eLife 2019; 8:e46472. [PMID: 31274109 PMCID: PMC6682400 DOI: 10.7554/elife.46472] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/04/2019] [Indexed: 12/17/2022] Open
Abstract
We identified a locus on mouse chromosome 10 that accounts for 60% of the genetic variance in methamphetamine intake in mice selectively bred for high versus low methamphetamine consumption. We nominated the trace amine-associated receptor 1 gene, Taar1, as the strongest candidate and identified regulation of the mu-opioid receptor 1 gene, Oprm1, as another contributor. This study exploited CRISPR-Cas9 to test the causal role of Taar1 in methamphetamine intake and a genetically-associated thermal response to methamphetamine. The methamphetamine-related traits were rescued, converting them to levels found in methamphetamine-avoiding animals. We used a family of recombinant inbred mouse strains for interval mapping and to examine independent and epistatic effects of Taar1 and Oprm1. Both methamphetamine intake and the thermal response mapped to Taar1 and the independent effect of Taar1 was dependent on genotype at Oprm1. Our findings encourage investigation of the contribution of Taar1 and Oprm1 variants to human methamphetamine addiction.
Collapse
Affiliation(s)
- Alexandra M Stafford
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
| | - Cheryl Reed
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
| | - Harue Baba
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
| | - Nicole AR Walter
- Division of NeuroscienceOregon National Primate Research CenterPortlandUnited States
| | - John RK Mootz
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
| | - Robert W Williams
- Department of Genetics, Genomics and InformaticsUniversity of Tennessee Health Sciences CenterMemphisUnited States
| | - Kim A Neve
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
- Veterans Affairs Portland Health Care SystemPortlandUnited States
| | - Lev M Fedorov
- Transgenic Mouse Models Shared Resource, Knight Cancer InstituteOregon Health & Science UniversityPortlandUnited States
| | - Aaron J Janowsky
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
- Veterans Affairs Portland Health Care SystemPortlandUnited States
- Department of PsychiatryOregon Health & Science UniversityPortlandUnited States
| | - Tamara J Phillips
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research CenterOregon Health & Science UniversityPortlandUnited States
- Veterans Affairs Portland Health Care SystemPortlandUnited States
| |
Collapse
|
5
|
Yazdani S, Salimi V, Eshraghian MR, Shayestehpour M, Pourtalebi-Firoozabadi A, Romani B, Shadab A, Aghasadeghi MR, Yavarian J. No genetic association between A118G polymorphism of μ-opioid receptor gene and schizophrenia and bipolar disorders. Indian J Psychiatry 2017; 59:483-486. [PMID: 29497192 PMCID: PMC5806329 DOI: 10.4103/psychiatry.indianjpsychiatry_53_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Schizophrenia (SZ) and bipolar disorder (BD) are chronic and multifactorial psychiatric disorders that might be affected by different genes in combination with environmental factors. There is evidence of association between polymorphisms of μ-opioid receptor gene (OPRM1) with these disorders. OBJECTIVES The aim of this study was to investigate the genetic association between OPRM1 A118G SNP in SZ and BD patients in comparison with healthy controls (HCs). MATERIALS AND METHODS One single-nucleotide polymorphism in OPRM1 was genotyped using TaqMan real-time PCR assay in 203 SZ and BD patients and 389 HCs. RESULTS There was no statistically significant difference in genotypic and allelic frequencies of OPRM1 A118G SNP between HCs and SZ/BD patients. CONCLUSIONS To find the underlying genetic factors associated with these complex disorders, further studies need to be conducted using larger sample size, different genetic populations, and different gene variations.
Collapse
Affiliation(s)
- Shaghayegh Yazdani
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Salimi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Eshraghian
- Department of Epidemiology and Biostatistics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Shayestehpour
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Pourtalebi-Firoozabadi
- Genomic Research Center, Birjand University of Medical Sciences, Birjand, Iran.,Department of Molecular Medicine, Birjand University of Medical Sciences, Birjand, Iran.,Department of Cellular and Molecular Medicine, Mehrvarzan-e-Saba Gostar Medical Rehabilitation and Maintenance Center of Chronic Mental Patients, Karaj, Alborz Province, Iran
| | - Bizhan Romani
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada.,Cellular and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Azadeh Shadab
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Jila Yavarian
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Phillips TJ, Mootz JRK, Reed C. Identification of Treatment Targets in a Genetic Mouse Model of Voluntary Methamphetamine Drinking. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 126:39-85. [PMID: 27055611 DOI: 10.1016/bs.irn.2016.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Methamphetamine has powerful stimulant and euphoric effects that are experienced as rewarding and encourage use. Methamphetamine addiction is associated with debilitating illnesses, destroyed relationships, child neglect, violence, and crime; but after many years of research, broadly effective medications have not been identified. Individual differences that may impact not only risk for developing a methamphetamine use disorder but also affect treatment response have not been fully considered. Human studies have identified candidate genes that may be relevant, but lack of control over drug history, the common use or coabuse of multiple addictive drugs, and restrictions on the types of data that can be collected in humans are barriers to progress. To overcome some of these issues, a genetic animal model comprised of lines of mice selectively bred for high and low voluntary methamphetamine intake was developed to identify risk and protective alleles for methamphetamine consumption, and identify therapeutic targets. The mu opioid receptor gene was supported as a target for genes within a top-ranked transcription factor network associated with level of methamphetamine intake. In addition, mice that consume high levels of methamphetamine were found to possess a nonfunctional form of the trace amine-associated receptor 1 (TAAR1). The Taar1 gene is within a mouse chromosome 10 quantitative trait locus for methamphetamine consumption, and TAAR1 function determines sensitivity to aversive effects of methamphetamine that may curb intake. The genes, gene interaction partners, and protein products identified in this genetic mouse model represent treatment target candidates for methamphetamine addiction.
Collapse
Affiliation(s)
- T J Phillips
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States; Veterans Affairs Portland Health Care System, Portland, OR, United States.
| | - J R K Mootz
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States
| | - C Reed
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
7
|
Liu J, Hu D, Jiang Y, Xi H, Li W. Association between single nucleotide polymorphisms in the OPRM1 gene and intraoperative remifentanil consumption in northern Chinese women. Pharmacology 2014; 94:273-9. [PMID: 25500932 DOI: 10.1159/000368082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/03/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND Recent studies have suggested that some single nucleotide polymorphisms (SNPs) in the human µ-opioid receptor gene (OPRM1) affect the postoperative analgesic efficacy of opioids and their side effects. In this study, we assessed the association between SNPs in the OPRM1 gene and intraoperative remifentanil consumption as well as perioperative side effects during gynecological hysteroscopic surgery in women from Northern China. METHODS We analyzed 178 women undergoing gynecological hysteroscopic surgery. SNP genotyping was performed using the SNaPshot method. The state anxiety index (SAI) and pressure pain threshold (PPT) of all patients were assessed preoperatively. Monitored anesthesia care was maintained by the intravenous infusion of remifentanil. Intraoperative remifentanil usage and perioperative side effects were recorded. Statistical analyses were performed using SPSS software. RESULTS Patients carrying one or two copies of the minor allele (G allele) of rs558025 required significantly more intraoperative remifentanil than patients without the minor allele (p = 0.001, corrected p = 0.006). There were no significant associations between the six SNPs and various clinical characteristics. No significant associations between the six SNPs and PPT or SAI were found in our study. CONCLUSIONS SNP rs558025 in the OPRM1 gene was associated with intraoperative remifentanil consumption during gynecological hysteroscopic surgery in our subjects.
Collapse
Affiliation(s)
- Jing Liu
- Department of Anesthesiology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | | | | | | | | |
Collapse
|
8
|
Eastwood EC, Phillips TJ. Morphine intake and the effects of naltrexone and buprenorphine on the acquisition of methamphetamine intake. GENES, BRAIN, AND BEHAVIOR 2014; 13:226-35. [PMID: 24152140 PMCID: PMC3976242 DOI: 10.1111/gbb.12100] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 09/23/2013] [Accepted: 10/21/2013] [Indexed: 11/29/2022]
Abstract
Some common genetic factors appear to influence risk for drug dependence across multiple drugs of abuse. In previous research, mice that were selectively bred for higher amounts of methamphetamine consumption, using a two-bottle choice methamphetamine drinking procedure, were found to be less sensitive to the locomotor stimulant effects of morphine and of the more selective μ-opioid receptor agonist fentanyl, compared to mice that were bred for low methamphetamine consumption. This suggested that μ-opioid receptor-mediated pathways may influence genetic risk for methamphetamine consumption. We hypothesized that these differences in opioid sensitivity would impact opioid intake in the methamphetamine drinking lines and that drugs with μ-opioid receptor activity would impact methamphetamine intake. Consumption of morphine was examined in 2, two-bottle choice studies, one that compared morphine to quinine consumption and another that used a saccharin fading procedure. Next, naltrexone (0, 0.5, 1, 2, 5, 10 and 20 mg/kg), a μ-opioid receptor antagonist, and buprenorphine (0, 1, 2 or 4 mg/kg), a μ-opioid receptor partial agonist, were each examined for their effects on the acquisition of methamphetamine consumption. Low methamphetamine drinking mice consumed more morphine compared to high methamphetamine drinking mice. Naltrexone did not alter methamphetamine consumption in either selected line; however, buprenorphine reduced methamphetamine intake in the high methamphetamine drinking line. These data show that greater sensitivity to opioids is associated with greater opioid intake and warrant further investigation of drugs with μ-opioid receptor-specific agonist activity in genetically determined differences in methamphetamine consumption.
Collapse
Affiliation(s)
- Emily C. Eastwood
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, 3181 SW Sam Jackson Rd., Portland, OR 97239, USA
| | - Tamara J. Phillips
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, 3181 SW Sam Jackson Rd., Portland, OR 97239, USA
- Veterans Affairs Medical Center, 3710 SW US Veterans Hospital Rd., Portland, Oregon, 97239, USA
| |
Collapse
|
9
|
Belknap JK, McWeeney S, Reed C, Burkhart-Kasch S, McKinnon CS, Li N, Baba H, Scibelli AC, Hitzemann R, Phillips TJ. Genetic factors involved in risk for methamphetamine intake and sensitization. Mamm Genome 2013; 24:446-58. [PMID: 24217691 PMCID: PMC3880562 DOI: 10.1007/s00335-013-9484-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 10/11/2013] [Indexed: 11/26/2022]
Abstract
Lines of mice were created by selective breeding for the purpose of identifying genetic mechanisms that influence the magnitude of the selected trait and to explore genetic correlations for additional traits thought to be influenced by shared mechanisms. DNA samples from high and low methamphetamine-drinking (MADR) and high and low methamphetamine-sensitization lines were used for quantitative trait locus (QTL) mapping. Significant additive genetic correlations between the two traits indicated a common genetic influence, and a QTL on chromosome X was detected for both traits, suggesting one source of this commonality. For MADR mice, a QTL on chromosome 10 accounted for more than 50 % of the genetic variance in that trait. Microarray gene expression analyses were performed for three brain regions for methamphetamine-naïve MADR line mice: nucleus accumbens, prefrontal cortex, and ventral midbrain. Many of the genes that were differentially expressed between the high and low MADR lines were shared in common across the three brain regions. A gene network highly enriched in transcription factor genes was identified as being relevant to genetically determined differences in methamphetamine intake. When the mu opioid receptor gene (Oprm1), located on chromosome 10 in the QTL region, was added to this top-ranked transcription factor network, it became a hub in the network. These data are consistent with previously published findings of opioid response and intake differences between the MADR lines and suggest that Oprm1, or a gene that impacts activity of the opioid system, plays a role in genetically determined differences in methamphetamine intake.
Collapse
Affiliation(s)
- John K. Belknap
- Veterans Affairs Medical Center, Oregon Health & Science University, Portland, OR, 97239 USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Shannon McWeeney
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
- Oregon Clinical and Translational Research Institute, Oregon Health & Science University, Portland, OR, 97239 USA
- Division of Biostatistics of Public Health & Preventative Medicine, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Cheryl Reed
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Sue Burkhart-Kasch
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Carrie S. McKinnon
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Na Li
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Harue Baba
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Angela C. Scibelli
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Robert Hitzemann
- Veterans Affairs Medical Center, Oregon Health & Science University, Portland, OR, 97239 USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| | - Tamara J. Phillips
- Veterans Affairs Medical Center, Oregon Health & Science University, Portland, OR, 97239 USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239 USA
- Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, 97239 USA
| |
Collapse
|
10
|
Ding S, Chen B, Zheng Y, Lu Q, Liu L, Zhuge QC. Association study of OPRM1 polymorphisms with Schizophrenia in Han Chinese population. BMC Psychiatry 2013; 13:107. [PMID: 23560613 PMCID: PMC3641981 DOI: 10.1186/1471-244x-13-107] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 03/12/2013] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The expression of μ-opioid receptor has important role in cognitive dysfunction in Schizophrenia (SZ). The results of studies about the association of polymorphisms of μ-opioid receptor gene (OPRM1) with SZ were inconsistent. METHODS We conducted a case-control study to investigate the genetic association between OPRM1 polymorphisms and SZ among the Han chinese population. 264 SZ patients and 264 age-matched control subjects were recruited. Four SNPs of OPRM1 were successfully genotyped by using PCR-RFLP. RESULTS Of four polymorphisms, rs1799971 and rs2075572 were shown to associate with SZ. Compared with the A allele of rs1799971 and C allele of rs2075572, the G allele of rs1799971 and rs2075572 was associated with an almost 0.46-fold risk (OR=0.46, 95% CI: 0.357-0.59, P<0.01) and 0.7-fold risk (OR=0.707, 95% CI: 0.534-0.937, P=0.015) of the occurrence of SZ,. When subjects were divided by gender, rs1799971 remained significant difference only in males (OR=0.309, 95% CI: 0.218-0.439 for G allele, P<0.01), and rs2075572 only in females (OR=0.399, 95% CI: 0.246-0.648 for G allele, P<0.01). In secondary analysis with subsets of patients, the G allele of rs1799971 (compared to the A allele) was associated with a decreased risk of all patients and male patients with apathy symptoms (OR=0.086, 95% CI: 0.048-0.151, P=0.01; OR=0.083, 95% CI: 0.045-0.153, P<0.01), and the G allele of rs2075572 (compared to the C allele) was associated with a decreased risk of all patients and female patients with positive family history (OR=0.468, 95% CI: 0.309-0.71, P<0.01; OR=0.34, 95% CI: 0.195-0.593, P<0.01). In addition, haplotype analysis revealed that two SNP haplotypes (A-C-C-G and G-C-C-A) were associated with decreased risks of SZ (P<0.01). The other two (G-C-C-G and G-G-C-G) with increased risks of SZ (P<0.01). CONCLUSIONS The present study demonstrated for the first time that the OPRM1 polymorphism may be a risk factor for schizophrenia in the Han Chinese. Further studies are needed to give a global view of this polymorphism in pathogenesis of schizophrenia in a large-scale sample, family-based association design or well-defined subgroups of schizophrenia.
Collapse
Affiliation(s)
- Saidan Ding
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, Department of Surgery, the First Affiliated Hospital of Wenzhou Medical College, Wenzhou, Zhejiang, China
| | - Bicheng Chen
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, Department of Surgery, the First Affiliated Hospital of Wenzhou Medical College, Wenzhou, Zhejiang, China
| | - Yong Zheng
- Clinical Laboratory, the Fifth People’s Hospital of Ruian city, Wenzhou, Zhejiang, China
| | - Qin Lu
- Neurosurgery Department, the First Affiliated Hospital of Wenzhou Medical College, Wenzhou, Zhejiang, China
| | - Leping Liu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, Department of Surgery, the First Affiliated Hospital of Wenzhou Medical College, Wenzhou, Zhejiang, China
| | - Qǐ -Chuan Zhuge
- Neurosurgery Department, the First Affiliated Hospital of Wenzhou Medical College, Wenzhou, Zhejiang, China
| |
Collapse
|
11
|
Gururajan A, Manning EE, Klug M, van den Buuse M. Drugs of abuse and increased risk of psychosis development. Aust N Z J Psychiatry 2012; 46:1120-35. [PMID: 22833579 DOI: 10.1177/0004867412455232] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE There is considerable evidence to suggest that the abuse of illicit drugs, particularly cannabis and methamphetamine, has aetiological roles in the pathogenesis of psychosis and schizophrenia. Factors that may increase susceptibility to the propsychotic effects of these drugs include the age at which the abuse starts as well as family history of genetic polymorphisms relevant to the pathophysiology of this disorder. However, the neurobiological mechanisms involved in drug abuse-associated psychosis remain largely unclear. METHODS AND RESULTS This paper presents an overview of the available evidence, including clinical, animal model, and molecular studies, with a focus on brain regions and neurotransmitters systems, such as dopamine and glutamate, previously implicated in psychosis. CONCLUSION It is clear that further studies are urgently needed to provide a greater insight into the mechanisms that mediate the long-term and neurodevelopmental effects of cannabis and methamphetamine. A dialogue between basic science and clinical research may help to identify at-risk individuals and novel pathways for treatment and prevention.
Collapse
Affiliation(s)
- Anand Gururajan
- Mental Health Research Institute, University of Melbourne, Melbourne, Australia
| | | | | | | |
Collapse
|
12
|
Carvalho M, Carmo H, Costa VM, Capela JP, Pontes H, Remião F, Carvalho F, Bastos MDL. Toxicity of amphetamines: an update. Arch Toxicol 2012; 86:1167-1231. [PMID: 22392347 DOI: 10.1007/s00204-012-0815-5] [Citation(s) in RCA: 267] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 02/02/2012] [Indexed: 01/06/2023]
Abstract
Amphetamines represent a class of psychotropic compounds, widely abused for their stimulant, euphoric, anorectic, and, in some cases, emphathogenic, entactogenic, and hallucinogenic properties. These compounds derive from the β-phenylethylamine core structure and are kinetically and dynamically characterized by easily crossing the blood-brain barrier, to resist brain biotransformation and to release monoamine neurotransmitters from nerve endings. Although amphetamines are widely acknowledged as synthetic drugs, of which amphetamine, methamphetamine, and 3,4-methylenedioxymethamphetamine (MDMA, ecstasy) are well-known examples, humans have used natural amphetamines for several millenniums, through the consumption of amphetamines produced in plants, namely cathinone (khat), obtained from the plant Catha edulis and ephedrine, obtained from various plants in the genus Ephedra. More recently, a wave of new amphetamines has emerged in the market, mainly constituted of cathinone derivatives, including mephedrone, methylone, methedrone, and buthylone, among others. Although intoxications by amphetamines continue to be common causes of emergency department and hospital admissions, it is frequent to find the sophism that amphetamine derivatives, namely those appearing more recently, are relatively safe. However, human intoxications by these drugs are increasingly being reported, with similar patterns compared to those previously seen with classical amphetamines. That is not surprising, considering the similar structures and mechanisms of action among the different amphetamines, conferring similar toxicokinetic and toxicological profiles to these compounds. The aim of the present review is to give an insight into the pharmacokinetics, general mechanisms of biological and toxicological actions, and the main target organs for the toxicity of amphetamines. Although there is still scarce knowledge from novel amphetamines to draw mechanistic insights, the long-studied classical amphetamines-amphetamine itself, as well as methamphetamine and MDMA, provide plenty of data that may be useful to predict toxicological outcome to improvident abusers and are for that reason the main focus of this review.
Collapse
Affiliation(s)
- Márcia Carvalho
- REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Levran O, Yuferov V, Kreek MJ. The genetics of the opioid system and specific drug addictions. Hum Genet 2012; 131:823-42. [PMID: 22547174 PMCID: PMC3721349 DOI: 10.1007/s00439-012-1172-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 04/15/2012] [Indexed: 12/21/2022]
Abstract
Addiction to drugs is a chronic, relapsing brain disease that has major medical, social, and economic complications. It has been established that genetic factors contribute to the vulnerability to develop drug addiction and to the effectiveness of its treatment. Identification of these factors may increase our understanding of the disorders, help in the development of new treatments and advance personalized medicine. In this review, we will describe the genetics of the major genes of the opioid system (opioid receptors and their endogenous ligands) in connection to addiction to opioids, cocaine, alcohol and methamphetamines. Particular emphasis is given to association and functional studies of specific variants. We will provide information on the sample populations and the size of each study, as well as a list of the variants implicated in association with addiction-related phenotypes, and with the effectiveness of pharmacotherapy for addiction.
Collapse
Affiliation(s)
- Orna Levran
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA.
| | | | | |
Collapse
|
14
|
Hart AB, de Wit H, Palmer AA. Genetic factors modulating the response to stimulant drugs in humans. Curr Top Behav Neurosci 2012; 12:537-77. [PMID: 22261702 PMCID: PMC3388157 DOI: 10.1007/7854_2011_187] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Individuals vary in their responses to stimulant drugs, and several lines of evidence suggest that the basis for this variation is at least partially genetic in origin. Association studies have examined the effects of polymorphisms in specific genes on acute and chronic responses to stimulant drugs. Several of these genetic polymorphisms are also associated with other psychiatric dimensions and disorders.This chapter examines the evidence for genetic associations between the genes that have been most carefully examined for their influence on the response to stimulant drugs.
Collapse
|
15
|
Abstract
The µ-opioid receptor is a primary target for clinically important opioid analgesics, including morphine, fentanyl and methadone. Many genetic variations have been identified in the human µ-opioid receptor MOP gene (OPRM1), and their implications have been reported in the effects of opioid drugs and susceptibility to drug dependence. Interestingly, agonistic and antagonistic opioid effects are inversely associated with the A118G polymorphism genotype. The A118G polymorphism may also be associated with substance dependence and susceptibility to other disorders, including epilepsy and schizophrenia. The IVS1+A21573G, IVS1-T17286C, and TAA+A5359G polymorphisms in the OPRM1 gene may be associated with alcohol, opioid and tobacco dependence, respectively. However, some studies have failed to confirm the correlations between the polymorphisms and opioid effects and substance dependence. Further studies are needed to elucidate the molecular mechanisms underlying the effects of OPRM1 polymorphisms.
Collapse
Affiliation(s)
| | - Kazutaka Ikeda
- Research Project for Addictive Substances, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| |
Collapse
|
16
|
Grant KM, LeVan TD, Wells SM, Li M, Stoltenberg SF, Gendelman HE, Carlo G, Bevins RA. Methamphetamine-associated psychosis. J Neuroimmune Pharmacol 2011; 7:113-39. [PMID: 21728034 PMCID: PMC3280383 DOI: 10.1007/s11481-011-9288-1] [Citation(s) in RCA: 160] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 06/21/2011] [Indexed: 01/03/2023]
Abstract
Methamphetamine (METH) is a frequent drug of abuse in U.S. populations and commonly associated with psychosis. This may be a factor in frequent criminal justice referrals and lengthy treatment required by METH users. Persecutory delusions and auditory hallucinations are the most consistent symptoms of METH-associated psychosis (MAP). MAP has largely been studied in Asian populations and risk factors have varied across studies. Duration, frequency and amount of use as well as sexual abuse, family history, other substance use, and co-occurring personality and mood disorders are risk factors for MAP. MAP may be unique with its long duration of psychosis and recurrence without relapse to METH. Seven candidate genes have been identified that may be associated with MAP. Six of these genes are also associated with susceptibility, symptoms, or treatment of schizophrenia and most are linked to glutamatergic neurotransmission. Animal studies of pre-pulse inhibition, attenuation of social interaction, and stereotypy and alterations in locomotion are used to study MAP in rodents. Employing various models, rodent studies have identified neuroanatomical and neurochemical changes associated with METH use. Throughout this review, we identify key gaps in our understanding of MAP and suggest potential directions for future research.
Collapse
Affiliation(s)
- Kathleen M Grant
- Department of Internal Medicine, VA Nebraska-Western Iowa Health Care System, University of Nebraska Medical Center, Omaha, NE 68198-5300, USA.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Dlugos AM, Hamidovic A, Hodgkinson C, Pei-Hong S, Goldman D, Palmer AA, de Wit H. OPRM1 gene variants modulate amphetamine-induced euphoria in humans. GENES, BRAIN, AND BEHAVIOR 2011; 10:199-209. [PMID: 21029375 PMCID: PMC3377371 DOI: 10.1111/j.1601-183x.2010.00655.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The μ-opioid receptor is involved in the rewarding effects of not only opioids like morphine but also psychostimulants like amphetamine. This study aimed to investigate associations between subjective response to amphetamine and genetic polymorphisms and haplotypes in the μ-opioid receptor including the exonic variant rs1799971 (Asp40Asn). One hundred and sixty-two Caucasian volunteers participated in three sessions receiving either placebo or d-amphetamine (10 and 20 mg). Associations between levels of self-reported Euphoria, Energy and Stimulation [Addiction Research Center Inventory 49-item questionnaire (ARCI-49)] after d-amphetamine ingestion and polymorphisms in OPRM1 were investigated. The intronic single nucleotide polymorphisms (SNPs) rs510769 and rs2281617 were associated with significantly higher ratings of Euphoria, Energy and Stimulation after 10 mg amphetamine. Feelings of Euphoria, Energy and Stimulation were also found to be associated with a two-SNP haplotype formed with rs1799971 and rs510769 and a three-SNP haplotype formed with rs1918760, rs2281617 and rs1998220. These results support the hypothesis that genetic variability in the μ-opioid receptor gene influences the subjective effects of amphetamine and may suggest new strategies for prevention and treatment of psychostimulant abuse.
Collapse
Affiliation(s)
- Andrea M. Dlugos
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, Illinois, USA
| | - Ajna Hamidovic
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, Illinois, USA
| | - Colin Hodgkinson
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Shen Pei-Hong
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Abraham A. Palmer
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, Illinois, USA
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, USA
| | - Harriet de Wit
- Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
18
|
Yamamoto H, Takamatsu Y, Imai K, Kamegaya E, Hagino Y, Watanabe M, Yamamoto T, Sora I, Koga H, Ikeda K. MOP Reduction During Long-Term Methamphetamine Withdrawal was Restored by Chronic Post-Treatment with Fluoxetine. Curr Neuropharmacol 2011; 9:73-8. [PMID: 21886566 PMCID: PMC3137205 DOI: 10.2174/157015911795017056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 04/17/2010] [Accepted: 05/26/2010] [Indexed: 11/22/2022] Open
Abstract
Previously, we found fluoxetine reduces methamphetamine preference in mice. However, effects of fluoxetine on developed methamphetamine preference and on methamphetamine induced gene expression changes have been largely unknown. The present study investigates effects of post-treatment with fluoxetine on methamphetamine dependence and on gene expressions after long-term withdrawal in mice. First, we examined whether chronic post-treatment with fluoxetine attenuated methamphetamine-conditioned place preference. Next, we examined the changes in gene expression levels after long-term withdrawal (with saline or fluoxetine treatment) following chronic methamphetamine treatment. Using mRNA from the pooled frontal cortices of 10 mice per group, gene expression analyses were performed using a custom-developed cDNA array and a real-time quantitative reverse transcription-PCR. Chronic post-treatments with fluoxetine abolished the conditioned place preference developed by methamphetamine administrations. Even after long-term withdrawal from repeated methamphetamine administration, µ-opioid receptor (MOP) gene expression was significantly reduced in the frontal cortex. The reduced MOP gene expression in the frontal cortex was restored by chronic administration with fluoxetine. These changes were confirmed by Western blot analyses. These findings suggest that the chronic post-treatments with fluoxetine might be effective for restoring the reduction of MOP levels in the frontal cortex following long-term abstinence from methamphetamine.
Collapse
Affiliation(s)
- H Yamamoto
- Division of Psychobiology, Tokyo Institute of Psychiatry, Tokyo, Japan
| | - Y Takamatsu
- Division of Psychobiology, Tokyo Institute of Psychiatry, Tokyo, Japan
| | - K Imai
- Laboratory for Medical Genomics, Department of Human Genome Technology, Kazusa DNA Research Institute, Chiba, Japan
| | - E Kamegaya
- Division of Psychobiology, Tokyo Institute of Psychiatry, Tokyo, Japan
| | - Y Hagino
- Division of Psychobiology, Tokyo Institute of Psychiatry, Tokyo, Japan
| | - M Watanabe
- Department of Anatomy, Hokkaido University School of Medicine, Sapporo, Japan
| | - T Yamamoto
- Division of Psychobiology, Tokyo Institute of Psychiatry, Tokyo, Japan
- Molecular Recognition, Yokohama City University, Yokohama, Japan
| | - I Sora
- Division of Psychobiology, Tokyo Institute of Psychiatry, Tokyo, Japan
- Department of Psychobiology, Tohoku University, School of Medicine, Sendai, Japan
| | - H Koga
- Laboratory for Medical Genomics, Department of Human Genome Technology, Kazusa DNA Research Institute, Chiba, Japan
| | - K Ikeda
- Division of Psychobiology, Tokyo Institute of Psychiatry, Tokyo, Japan
| |
Collapse
|
19
|
Mague SD, Blendy JA. OPRM1 SNP (A118G): involvement in disease development, treatment response, and animal models. Drug Alcohol Depend 2010; 108:172-82. [PMID: 20074870 PMCID: PMC2887703 DOI: 10.1016/j.drugalcdep.2009.12.016] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Revised: 12/17/2009] [Accepted: 12/17/2009] [Indexed: 12/21/2022]
Abstract
Endogenous opioids acting at mu-opioid receptors mediate many biological functions. Pharmacological intervention at these receptors has greatly aided in the treatment of acute and chronic pain, in addition to other uses. However, the development of tolerance and dependence has made it difficult to adequately prescribe these therapeutics. A common single nucleotide polymorphism (SNP), A118G, in the mu-opioid receptor gene can affect opioid function and, consequently, has been suggested to contribute to individual variability in pain management and drug addiction. Investigation into the role of A118G in human disease and treatment response has generated a large number of association studies across various disease states as well as physiological responses. However, characterizing the functional consequences of this SNP and establishing if it causes or contributes to disease phenotypes have been significant challenges. In this manuscript, we will review a number of association studies as well as investigations of the functional impact of this gene variant. In addition, we will describe a novel mouse model that was generated to recapitulate this SNP in mice. Evaluation of models that incorporate known human genetic variants into a tractable system, like the mouse, will facilitate the understanding of discrete contributions of SNPs to human disease.
Collapse
Affiliation(s)
| | - Julie A. Blendy
- Address correspondence to: Julie A. Blendy, Ph.D., Department of Pharmacology, University of Pennsylvania, TRL, 125 South 31st Street, Tel: (215) 898-0730, Fax: (215) 573-2041,
| |
Collapse
|
20
|
Nishizawa D, Hayashida M, Nagashima M, Koga H, Ikeda K. Genetic polymorphisms and human sensitivity to opioid analgesics. Methods Mol Biol 2010; 617:395-420. [PMID: 20336437 DOI: 10.1007/978-1-60327-323-7_29] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Opioid analgesics are commonly used for the treatment of acute as well as chronic, moderate to severe pain. Well-known, however, is the wide interindividual variability in sensitivity to opioids that exists, which has often been a critical problem in pain treatment. To date, only a limited number of studies have addressed the relationship between human genetic variations and sensitivity to opioids, and such studies are still in their early stages. Therefore, revealing the relationship between genetic variations in many candidate genes and individual differences in sensitivity to opioids will provide valuable information for appropriate individualization of opioid doses required for adequate pain control. Although the methodologies for such association studies can be diverse, here we summarize protocols for investigating the association between genetic polymorphisms and sensitivity to opioids in human volunteers and patients undergoing painful surgery.
Collapse
Affiliation(s)
- Daisuke Nishizawa
- Division of Psychobiology, Tokyo Institute of Psychiatry, Tokyo, Japan
| | | | | | | | | |
Collapse
|
21
|
Bousman CA, Glatt SJ, Everall IP, Tsuang MT. Genetic association studies of methamphetamine use disorders: A systematic review and synthesis. Am J Med Genet B Neuropsychiatr Genet 2009; 150B:1025-49. [PMID: 19219857 DOI: 10.1002/ajmg.b.30936] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Efforts to understand the biological processes that increase susceptibility to methamphetamine (METH) use disorders (i.e., abuse, dependence, and psychosis) have uncovered several putative genotypic variants. However, to date a synthesis of this information has not been conducted. Thus, systematic searches of the current literature were undertaken for genetic-association studies of METH use disorders. Each gene's chromosomal location, function, and examined polymorphic markers were extracted. Frequencies, odds ratios and 95% confidence intervals for risk alleles, as well as sample size and power, were calculated. We uncovered 38 studies examining 39 genes, of which 18 were found to have a significant genotypic, allelic, and/or haplotypic association with METH use disorders. Three genes (COMT, DRD4, and GABRA1) were associated with METH abuse, nine (ARRB2, BDNF, CYP2D6, GLYT1, GSTM1, GSTP1, PDYN, PICK1, and SLC22A3) with METH dependence, two (AKT1 and GABRG2) with METH abuse/dependence, and four (DTNBP1, OPRM1, SNCA, and SOD2) with METH psychosis. Limitations related to phenotypic classification, statistical power, and potential publication bias in the current literature were noted. Similar to other behavioral, psychiatric, and substance use disorders, the genetic epidemiology of METH use disorders is complex and likely polygenic. National and international collaborative efforts are needed to increase the availability of large population-based samples and improve upon the power to detect genetic associations of small magnitude. Further, replication of the findings reviewed here along with further development of more rigorous methodologies and reporting protocols will aid in delineating the complex genetic epidemiology of METH use disorders.
Collapse
Affiliation(s)
- Chad A Bousman
- Department of Psychiatry, Center for Behavioral Genomics, University of California San Diego, La Jolla, 92037, USA
| | | | | | | |
Collapse
|
22
|
Association between OPRM1 gene polymorphisms and fentanyl sensitivity in patients undergoing painful cosmetic surgery. Pain 2009; 147:194-201. [DOI: 10.1016/j.pain.2009.09.004] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 09/01/2009] [Accepted: 09/04/2009] [Indexed: 11/21/2022]
|
23
|
Kotaka T, Ujike H, Okahisa Y, Takaki M, Nakata K, Kodama M, Inada T, Yamada M, Uchimura N, Iwata N, Sora I, Iyo M, Ozaki N, Kuroda S. G72 gene is associated with susceptibility to methamphetamine psychosis. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:1046-9. [PMID: 19482054 DOI: 10.1016/j.pnpbp.2009.05.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 05/18/2009] [Accepted: 05/25/2009] [Indexed: 12/21/2022]
Abstract
Methamphetamine psychosis is considered as one of the pharmacological models of schizophrenia, and a hyperdopaminergic one. However, many lines of experimental evidence indicate that glutamatergic signaling is also involved in development of methamphetamine psychosis. Several genes related to glutamate function, e.g. the DTNBP1, G72, and GRM3 genes, were shown to be associated with schizophrenia susceptibility. Recently, we found significant association of the DTNBP1 gene with methamphetamine psychosis. This finding prompted us to examine the G72 gene encoding the d-amino acid oxidase activator (DAOA), which metabolizes d-serine, an NMDA co-agonist, in methamphetamine psychosis. Six SNPs of the G72 gene, which previously showed significant association with schizophrenia, were analyzed in 209 patients with methamphetamine psychosis and 291 age- and sex-matched normal controls. One SNP of M22 (rs778293) showed a significant association with methamphetamine psychosis (genotype: p=0.00016, allele: p=0.0015). Two haplotypes G-A of M12 (rs3916965)-M15 (rs2391191) (p=0.00024) and T-T of M23 (rs947267)-M24 (rs1421292) (p=0.00085) also showed associations with methamphetamine psychosis. The present findings suggest that the G72 gene may contribute to a predisposition to not only schizophrenia but also to methamphetamine psychosis.
Collapse
Affiliation(s)
- Tatsuya Kotaka
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama 700-8558, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ono T, Muto A, Kaneda T, Arita E, Yoshida T. Novel linkage disequilibrium of single nucleotide polymorphisms in the transcriptional regulatory region of micro-opioid receptor gene in Japanese population. Biol Pharm Bull 2009; 32:721-3. [PMID: 19336912 DOI: 10.1248/bpb.32.721] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
micro-Opioid receptor agonists, such as morphine, are widely applied in pain therapy clinical practice. However, the effects exerted by morphine via receptor are influenced by individual specificity. Single nucleotide polymorphisms (SNPs) in micro-opioid receptor gene (OPRM1) have been reported to influence receptor expression and function. Subsequently, we analyzed SNPs frequency and linkage disequilibrium associated with OPRM1 transcriptional region and 4 exons among healthy Japanese individuals. Consequently, we detected 10 SNPs (-1748G/A, -1565T/C, -1045A/G, -172G/T, -38C/A, 118A/G, ivs2+31 G/A, ivs2+691 C/G, ivs4+274 A/G, and ivs4+435 G/A). Moreover, linkage analysis revealed novel linkage between -1748G/A and -172G/T, which was not observed in studies performed in other nations. In contrast, SNPs frequency detected in this study was similar to previously reported results on Asians; however, linkage disequilibrium reports from different nations differed. These results possibly provide useful information for OPRM1 genotyping in the Japanese population.
Collapse
Affiliation(s)
- Takeshi Ono
- Department of Pathophysiology, Faculty of Pharmaceutical Sciences, Hoshi University, Japan
| | | | | | | | | |
Collapse
|
25
|
Haile CN, Kosten TR, Kosten TA. Pharmacogenetic treatments for drug addiction: cocaine, amphetamine and methamphetamine. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2009; 35:161-77. [PMID: 19462300 PMCID: PMC2754046 DOI: 10.1080/00952990902825447] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Pharmacogenetics uses genetic variation to predict individual differences in response to medications and holds much promise to improve treatment of addictive disorders. OBJECTIVES To review how genetic variation affects responses to cocaine, amphetamine, and methamphetamine and how this information may guide pharmacotherapy. METHODS We performed a cross-referenced literature search on pharmacogenetics, cocaine, amphetamine, and methamphetamine. RESULTS We describe functional genetic variants for enzymes dopamine-beta-hydroxylase (DbetaH), catechol-O-methyltransferase (COMT), and dopamine transporter (DAT1), dopamine D4 receptor, and brain-derived neurotrophic factor (BDNF). A single nucleotide polymorphism (SNP; C-1021T) in the DbetaH gene is relevant to paranoia associated with disulfiram pharmacotherapy for cocaine addiction. Individuals with variable number tandem repeats (VNTR) of the SLC6A3 gene 3'-untranslated region polymorphism of DAT1 have altered responses to drugs. The 10/10 repeat respond poorly to methylphenidate pharmacotherapy and the 9/9 DAT1 variant show blunted euphoria and physiological response to amphetamine. COMT, D4 receptor, and BDNF polymorphisms are linked to methamphetamine abuse and psychosis. CONCLUSIONS Disulfiram and methylphenidate pharmacotherapies for cocaine addiction are optimized by considering polymorphisms affecting DbetaH and DAT1 respectively. Altered subjective effects for amphetamine in DAT1 VNTR variants suggest a 'protected' phenotype. SCIENTIFIC SIGNIFICANCE Pharmacogenetic-based treatments for psychostimulant addiction are critical for successful treatment.
Collapse
Affiliation(s)
- Colin N Haile
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, and Michael E DeBakey VA Medical Center, Houston, Texas 77030, USA
| | | | | |
Collapse
|
26
|
Hayashida M, Nagashima M, Satoh Y, Katoh R, Tagami M, Ide S, Kasai S, Nishizawa D, Ogai Y, Hasegawa J, Komatsu H, Sora I, Fukuda K, Koga H, Hanaoka K, Ikeda K. Analgesic requirements after major abdominal surgery are associated with OPRM1 gene polymorphism genotype and haplotype. Pharmacogenomics 2008; 9:1605-16. [DOI: 10.2217/14622416.9.11.1605] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: The association between SNPs of the human OPRM1 gene encoding the µ-opioid receptor and postoperative analgesic requirements in surgical patients remains controversial. Here, we evaluate whether any of the five tag SNPs (A118G, IVS2+G691C, IVS3+G5953A, IVS3+A8449G and TAA+A2109G) representing the four linkage disequilibrium blocks of the OPRM1 gene influences postoperative analgesic requirements. Materials & methods: We studied 138 adult Japanese patients who underwent major open abdominal surgery under combined general and epidural anesthesia and received continuous postoperative epidural analgesia with opioids. Results: The 118G homozygous (GG) patients required 24-h postoperative analgesics more than 118A homozygous (AA) and heterozygous (AG) patients. Tag SNP haplotypes also were associated with 24-h postoperative analgesic requirements. Conclusions: These results suggest that OPRM1 gene tag SNP genotypes and haplotypes can primarily contribute to prediction of postoperative analgesic requirements in individual patients undergoing major open abdominal surgery.
Collapse
Affiliation(s)
- Masakazu Hayashida
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Makoto Nagashima
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Yasuo Satoh
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Ryoji Katoh
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Megumi Tagami
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Soichiro Ide
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Shinya Kasai
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Daisuke Nishizawa
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Yasukazu Ogai
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Junko Hasegawa
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Hiroshi Komatsu
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Ichiro Sora
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Kenichi Fukuda
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Hisashi Koga
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Kazuo Hanaoka
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| | - Kazutaka Ikeda
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156–8585, Japan
| |
Collapse
|
27
|
Ehlers CL, Lind PA, Wilhelmsen KC. Association between single nucleotide polymorphisms in the mu opioid receptor gene (OPRM1) and self-reported responses to alcohol in American Indians. BMC MEDICAL GENETICS 2008; 9:35. [PMID: 18433502 PMCID: PMC2386778 DOI: 10.1186/1471-2350-9-35] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Accepted: 04/23/2008] [Indexed: 11/10/2022]
Abstract
BACKGROUND Variation in response to the hedonic and adverse effects of a substance is in part an inherited factor that may influence its use, abuse and dependence. The mu opioid receptor is the primary site of action for opiates and individuals with polymorphisms in this receptor appear to have variation in the CNS effects of opiates. Several studies have suggested that this receptor may also mediate some of the effects of non-opioid drugs, such as alcohol. The purpose of this study was to investigate associations between 13 single nucleotide polymorphisms in the mu opioid receptor gene (OPRM1) with self-reported responses to alcohol, an endophenotype associated with the development of alcohol dependence, in American Indians living on eight contiguous reservations. METHODS Each participant gave a blood sample and completed a structured diagnostic interview. Additionally, response to alcohol was indexed using the expectation version of the subjective high assessment scale (SHAS-E). SNPs were genotyped in 251 participants and data analyses were conducted using SOLAR. RESULTS The estimated heritability (h2) for the SHAS-E phenotypes ranged from 0.01 to 0.28. Endorsing the expectation of a more intense response on one or more of the following items from the SHAS-E: buzzed, clumsy, dizzy, drunk, effects, high, nausea, sleepy, talkative, terrible, and/or uncomfortable after imbibing 2-3 drinks was significantly associated with having at least one minor allele for at least one of 7 SNPs (p < 0.01) in the OPRM1 receptor gene. CONCLUSION These studies provide data to suggest that the minor allele, for most of the polymorphisms in the OPRM1 receptor gene investigated, was found to be associated with a more intense, and/or more adverse, response to alcohol, traits that are significantly correlated with lowered quantity of alcohol consumption and less susceptibility to dependence in this Indian population. These data further suggest that making conclusions on the role of the mu opiod receptor gene in the development of alcohol dependence may be limited if only one polymorphism in the gene is evaluated in isolation.
Collapse
Affiliation(s)
- Cindy L Ehlers
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| | | | | |
Collapse
|
28
|
Hishimoto A, Cui H, Mouri K, Nushida H, Ueno Y, Maeda K, Shirakawa O. A functional polymorphism of the µ-opioid receptor gene is associated with completed suicides. J Neural Transm (Vienna) 2008; 115:531-6. [DOI: 10.1007/s00702-007-0853-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Accepted: 10/30/2007] [Indexed: 12/01/2022]
|
29
|
Otani K, Ujike H, Sakai A, Okahisa Y, Kotaka T, Inada T, Harano M, Komiyama T, Hori T, Yamada M, Sekine Y, Iwata N, Iyo M, Sora I, Ozaki N, Kuroda S. Reduced CYP2D6 activity is a negative risk factor for methamphetamine dependence. Neurosci Lett 2008; 434:88-92. [PMID: 18280655 DOI: 10.1016/j.neulet.2008.01.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 01/12/2008] [Accepted: 01/15/2008] [Indexed: 11/26/2022]
Abstract
Because methamphetamine (METH) is metabolized by CYP2D6 at the first step of hydroxylation and demethylation, it is possible that functional variants of CYP2D6 alter susceptibility to methamphetamine-induced dependence. We genotyped CYP2D6*1, *4, *5, *10, and *14 for 202 patients with METH dependence and 337 controls in a Japanese population and found a significant association of the CYP2D6 gene with METH dependence (p=0.0299). The patients had fewer *10 and *14 alleles, which are hypofunction alleles, than the controls. CYP2D6 genotypes were divided into three phenotypes: extensive metabolizers, intermediate metabolizers, and poor metabolizers. There was no poor metabolizer among our Japanese subjects, and intermediate metabolizers of CYP2D6 were significantly fewer in methamphetamine-dependent subjects than in controls (p=0.0212), with an odds ratio of 0.62 (95% confidence interval: 0.51-0.76). The present study demonstrated that reduced CYP2D6 activity was a negative risk factor for methamphetamine dependence.
Collapse
Affiliation(s)
- Kyohei Otani
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Shikata-cho 2-5-1, Okayama 700-8558, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Amfetamine abuse has grown into a worldwide epidemic. Methamfetamine, a derivative of amfetamine made from readily accessible chemicals, has plagued the US since the 1960s, with an alarming recent surge in the numbers of those meeting the criteria for amfetamine abuse and dependence. We review this problem using a computerised literature search (PubMed 1964-2007) to summarise knowledge from animal and human studies about treatments for amfetamine dependence, while exploring the potential of pharmacogenetics to help uncover new treatment targets. Several promising therapeutic targets have come from animal models of reward, drug-taking behaviour and withdrawal. Although preclinical and selected clinical results have been promising, clinical studies have yielded inconsistent results. To improve these outcomes, pharmacogenetic studies may be used to identify candidate alleles that predict therapeutic response. Exciting preclinical findings and a steady progression of clinical results offer hope for the development of a treatment for amfetamine dependence.
Collapse
Affiliation(s)
- Kevin P Hill
- Alcohol and Drug Abuse Treatment Program, McLean Hospital, Belmont, Massachusetts 02478-9106, USA.
| | | |
Collapse
|
31
|
Abstract
This paper is the 29th consecutive installment of the annual review of research concerning the endogenous opioid system, now spanning 30 years of research. It summarizes papers published during 2006 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurological disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, CUNY, 65-30 Kissena Blvd., Flushing, NY 11367, United States.
| |
Collapse
|
32
|
Candidate gene polymorphisms predicting individual sensitivity to opioids. Naunyn Schmiedebergs Arch Pharmacol 2007; 377:269-81. [DOI: 10.1007/s00210-007-0205-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Accepted: 10/18/2007] [Indexed: 11/26/2022]
|
33
|
Nagashima M, Katoh R, Sato Y, Tagami M, Kasai S, Ikeda K. Is there genetic polymorphism evidence for individual human sensitivity to opiates? Curr Pain Headache Rep 2007; 11:115-23. [PMID: 17367590 DOI: 10.1007/s11916-007-0008-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Opiate analgesics have been widely used for severe acute pain and chronic cancer-related pain. Individual differences in the effectiveness of opiates and their side effects limit the clinical benefits and increase risks of drug abuse. Genetic factors might affect variations of opiate sensitivity. The mu opioid peptide receptor (MOP) is the principal site of pharmacologic actions for most clinically important opiate drugs. Recent studies using various knockout mice and recombinant-inbred strain CXBK mice have indicated that the analgesic effect of morphine is dependent on the amount of the MOP. There are more than 100 polymorphisms identified in the human MOP (OPRM1) gene. These polymorphisms might be correlated with OPRM1 mRNA stability and opiate sensitivity, including opiate analgesia, tolerance, and dependence. More precise studies on the relationship between gene polymorphisms and opiate sensitivity will enable realization of personalized pain treatment by predicting opiate sensitivity and requirement for each patient.
Collapse
Affiliation(s)
- Makoto Nagashima
- Department of Molecular Psychiatry, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156-8585, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Aoyama N, Takahashi N, Kitaichi K, Ishihara R, Saito S, Maeno N, Ji X, Takagi K, Sekine Y, Iyo M, Harano M, Komiyama T, Yamada M, Sora I, Ujike H, Iwata N, Inada T, Ozaki N. Association between gene polymorphisms of SLC22A3 and methamphetamine use disorder. Alcohol Clin Exp Res 2006; 30:1644-9. [PMID: 17010131 DOI: 10.1111/j.1530-0277.2006.00215.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Methamphetamine (MAP) is one of the most frequently used illegal substances in Japan, and family and twin studies have suggested that genetic factors contribute to psychostimulant dependence, including MAP dependence. Organic cation transporter 3 (OCT3) has been reported to be involved in the disposition of MAP as well as MAP-induced behavioral changes in animals. Moreover, SLC22A3 (which encodes OCT3) is a candidate gene for MAP dependence because it is located within a chromosomal region associated with substance dependence. METHODS Using 96 healthy control subjects, linkage disequilibrium (LD) within the SLC22A3 was investigated, and 5 single-nucleotide polymorphisms (SNPs) were selected as haplotype tag SNPs to search for an association with MAP dependence. Single-marker analyses and haplotype analyses of these SNPs were performed in 213 subjects with MAP dependence and 443 healthy controls. RESULTS SLC22A3 polymorphisms were not significantly associated with MAP dependence in any of the single-marker and haplotype analyses. When subjects with MAP dependence were divided into polysubstance and single-MAP users, genotype and allele frequency of SNP2 (p=0.024, p=0.011, respectively), allele frequency of SNP3 (p=0.037), and haplotypic frequencies for these 2 SNPs (p=0.0438) differed significantly between groups. CONCLUSIONS These results suggest that polymorphisms of SLC22A3 are related to the development of polysubstance use in Japanese patients with MAP dependence.
Collapse
Affiliation(s)
- Nagisa Aoyama
- Department of Medical Technology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|