751
|
Saito M, Ehringer MA, Toth R, Oros M, Szakall I, Sikela JM, Vadasz C. Variants of kappa-opioid receptor gene and mRNA in alcohol-preferring and alcohol-avoiding mice. Alcohol 2003; 29:39-49. [PMID: 12657375 DOI: 10.1016/s0741-8329(02)00322-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Results of recent studies have indicated an association between voluntary alcohol intake and activities of kappa-opioid receptor systems in animal models. We assessed the possibility that genetic differences observed in alcohol preference among mouse strains are related to possible polymorphisms of the kappa-opioid receptor gene (Oprk1). We compared DNA sequences of the coding region and the promoter/regulatory region of Oprk1 among C57BL/6ByJ (B6, alcohol-preferring), BALB/cJ (alcohol-avoiding), CXBI (alcohol-avoiding), and six B6.C and B6.I Recombinant QTL Introgression (RQI) strains, which carry approximately 3% of the donor BALB/cJ genome in the background B6 genome and showed various alcohol preferences. Although there were no sequence differences in the coding region, BALB/cJ had a single nucleotide polymorphism (SNP) in the promoter region, which was not detected in other strains. The results indicate that the difference in alcohol preference between B6 and BALB/cJ is not correlated with polymorphisms of Oprk1. However, results of further studies comparing Oprk1 mRNA expression between B6 and BALB/cJ showed that Oprk1 expression is regulated differently in these strains. Also, DBA/2J mice (alcohol-avoiding) showed expression of Oprk1 mRNA subtypes (alternatively spliced) different from B6 and BALB/cJ mice. Search of the Celera Genomics database indicated that DBA/2J had several SNP sites in the promoter/regulatory regions, which might explain the different expression of Oprk1 mRNA subtypes in this strain. The strain-dependent variation in the expression of alternatively spliced genes can be a significant source of phenotypic variation of complex traits such as alcohol preference.
Collapse
MESH Headings
- Alcohol Drinking/genetics
- Animals
- Avoidance Learning/physiology
- Base Sequence
- Female
- Genetic Variation/genetics
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Knockout
- Molecular Sequence Data
- Polymorphism, Genetic
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Receptors, Opioid, kappa/biosynthesis
- Receptors, Opioid, kappa/deficiency
- Receptors, Opioid, kappa/genetics
- Sequence Homology, Nucleic Acid
- Species Specificity
Collapse
Affiliation(s)
- Mariko Saito
- Laboratory of Neurobehavior Genetics, The Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, NY 10962, USA.
| | | | | | | | | | | | | |
Collapse
|
752
|
Su ZG, Zhang SZ, Hou YP, Li T, Nebert DW, Zhang L, Huang DJ, Liao LC, Xiao CY. Single-nucleotide polymorphisms in the lipoprotein lipase gene associated with coronary heart disease in Chinese. Eur J Pharmacol 2002; 454:9-18. [PMID: 12408999 DOI: 10.1016/s0014-2999(02)02427-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Coronary heart disease is a complex disease reflecting the interaction of multiple genes with the environment (e.g. diet, life style). Lipoprotein lipase (LPL) plays an important role in lipid metabolism and the pathogenesis of coronary atherosclerosis. Recent associations between single-nucleotide polymorphisms in the LPL gene and heart disease have been reported, but little is known in Chinese. The LPL gene spans >26 kb, with an mRNA of 3549 bp. In the present study, we screened 5155 bp (565 bp of 5' flanking region, nine exons and donor- and acceptor-splice sites, and some intronic bases) in 160 Chinese patients with confirmed coronary heart disease and 150 age- and gender-matched controls. Thirteen of the sixteen single-nucleotide polymorphisms that we found have not been previously reported. In males, significant (P<0.05) differences between the coronary heart disease patients and controls were found for five single-nucleotide polymorphisms: -421G>A (5' flanking region); +13,577C>A (intron 2); +16,052G>A, R192Q (exon 5); +16,173C>G and +16,177T>C (intron 5). In females, significant differences between the patients with coronary heart disease and controls were found for only the -421G>A and +16,052G>A (R192Q) mutations. Among the coronary heart disease males, significant (P<0.05) associations were found between the low-HDL high-triglyceride (LHDL/HTG) phenotype and the non-LHDL/HTG trait for the 5' flanking-421G, the intron 2+13,577C, and the exon 5+16,052G mutations, with odds-ratios (ORs)[confidence intervals] of 3.90[1.12-13.66], 3.38[1.22-9.40], and 3.22[1.04-10.01], respectively; no corresponding associations were found in females. There were 69, 51, 57 and 41 unphased haplotype patterns in male coronary heart disease, male control, female coronary heart disease and female control groups, respectively; the computer program PM-Plus found the heterogeneity model by far the best fit (P<0.0001 in males, >0.01 in females). These data show that some single-nucleotide polymorphisms in the LPL gene among Chinese are associated with abnormal lipid and lipoprotein profiles and predisposition to coronary heart disease, a genetically heterogeneous complex disease, and that they are gender-specific.
Collapse
Affiliation(s)
- Zhi G Su
- Department of Medical Genetics, West China Hospital, Sichuan University, Sichuan, Chengdu 610041, China
| | | | | | | | | | | | | | | | | |
Collapse
|
753
|
Xin L, Wang ZJ. Bioinformatic analysis of the human mu opioid receptor (OPRM1) splice and polymorphic variants. AAPS PHARMSCI 2002; 4:E23. [PMID: 12645995 PMCID: PMC2751312 DOI: 10.1208/ps040423] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Mu opioid receptor (OPRM1), a member of the G-protein coupled receptor superfamily, mediates the analgesic and euphoric effects of opioid drugs. The sequences of OPRM1 cDNA and reported splice variants were used to search the public and Celera genomic databases. The matched sequences were analyzed to assemble an OPRM1 genomic contig. Human OPRM1 gene was estimated to span at least 90 kb in the chromosome 6q24-25 region. Four coding exons are separated by 3 introns. While intron 2 has only 773 bp, these databases for the first time provide the precise length of and other information about long introns 1 and 3, containing 50 and 27 kb, respectively. When a consensus exon/intron splice junction at the end of the coding exon 3 was not utilized, it may have resulted in continuous translation of the exon to yield the splice variant OPRM1A. The study did not identify human orthologs of other OPRM1 variants that had been reported for mouse OPRM1, although several proposed exons were found to be included in mouse genomic clones. Single nucleotide polymorphisms in the OPRM1 gene were also analyzed and summarized, which could provide potential polymorphic markers for molecular genetic studies.
Collapse
Affiliation(s)
- Lili Xin
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, 60612 Chicago, IL
| | - Zaijie Jim Wang
- Department of Biopharmaceutical Sciences, College of Pharmacy, University of Illinois, 60612 Chicago, IL
| |
Collapse
|
754
|
Abstract
Pharmacogenetic research dedicated to the investigation of inherited factors that influence drug response has produced exciting results over the past decade. Adding to the knowledge that genetic variation in metabolic enzymes may cause drug-related adverse reactions, recent studies indicate that variation in neurotransmitter receptors can also be the cause of treatment failure. In addition, recent studies have attempted to use genetic information for the prediction of treatment outcome. The aim of this review is to summarize the most significant findings in pharmacogenetic research in relation to CNS drugs and to outline how these studies could lead to the individualization of drug treatment.
Collapse
Affiliation(s)
- Sanober Shaikh
- Clinical Neuropharmacology, Department of Psychological Medicine, Institute of Psychiatry, London, UK
| | | |
Collapse
|
755
|
Abstract
Addiction to drugs, such as heroin, cocaine and alcohol, exacts great human and financial costs on society, but the development of pharmacotherapies for addiction has been largely neglected by the pharmaceutical industry. With advances in our understanding of the underlying biology of addictions now opening the door for the development of novel pharmacotherapies, it could be time for a reassessment of involvement in this increasingly important therapeutic area. Here, we summarize the current approved and implemented pharmacotherapeutic approaches to the treatment of addiction, and then highlight the most promising areas for future drug development from the perspective of our laboratory and our National Institutes of Health (NIH) National Institute on Drug Abuse (NIDA) Research Center.
Collapse
Affiliation(s)
- Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, 1230 York Avenue, New York, New York 10021, USA.
| | | | | |
Collapse
|
756
|
Jorm AF, Prior M, Sanson A, Smart D, Zhang Y, Tan S, Easteal S. Lack of association of a single-nucleotide polymorphism of the mu-opioid receptor gene with anxiety-related traits: results from a cross-sectional study of adults and a longitudinal study of children. AMERICAN JOURNAL OF MEDICAL GENETICS 2002; 114:659-64. [PMID: 12210283 DOI: 10.1002/ajmg.10643] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
There is evidence from animal experiments that the mu- and delta-opioid receptors may play a role in anxiety and depression. It might therefore be expected that functional polymorphisms of these genes in humans are associated with anxiety and depression. We investigated a single-nucleotide polymorphism (Asn40Asp) of the mu-opioid receptor gene (OPRM1). This association was investigated in two samples: 1) a cross-sectional survey of 867 community-living adults aged 18-79 years who were assessed for anxiety and depression symptoms and related personality traits; and 2) a longitudinal study of childhood temperament in which 660 children were followed from infancy to the mid-teens and assessed for anxiety-related temperament and behavior problems. The data did not support a role for the Asn40Asp polymorphism in anxiety and depression, despite adequate statistical power to detect small effects.
Collapse
Affiliation(s)
- Anthony F Jorm
- Centre for Mental Health Research, Australian National University, Canberra, Australia.
| | | | | | | | | | | | | |
Collapse
|
757
|
Kellogg SH, Ho A, Bell K, Schluger RP, McHugh PF, McClary KA, Kreek MJ. The Personality Assessment Inventory Drug Problems Scale: a validity analysis. J Pers Assess 2002; 79:73-84. [PMID: 12227669 DOI: 10.1207/s15327752jpa7901_05] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
An analysis of the relationship among the Personality Assessment Inventory (PAI; Morey, 1991, 1996) Drug Problems (DRG) scale scores, the Addiction Severity Index (ASI; McLellan et al., 1992) scores, and urine toxicology reports revealed that the PAI Drug Problem scale scores of 100 substance-using and substance-abusing men and women were distributed in a manner that was in agreement with the guidelines suggested by Morey (1991, 1996) in the PAI manual. There were significant correlations among the PAI DRG scale and the ASI scales related to frequency of use, negative consequences of use, and need and desire for treatment. Overall, higher scores did reflect both more serious involvements with drug use and more serious problems as a consequence of their involvement.
Collapse
Affiliation(s)
- Scott H Kellogg
- Laboratory of the Biology of the Addictive Diseases, Rockefeller University, New York, NY 10021-6399, USA.
| | | | | | | | | | | | | |
Collapse
|
758
|
Xu K, Liu XH, Nagarajan S, Gu XY, Goldman D. Relationship of the delta-opioid receptor gene to heroin abuse in a large Chinese case/control sample. AMERICAN JOURNAL OF MEDICAL GENETICS 2002; 110:45-50. [PMID: 12116270 DOI: 10.1002/ajmg.10374] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Pharmacological and electrophysiological evidence has shown that opioid receptors are involved in the mechanism of heroin dependence. Thus, opioid receptors are appropriate candidate genes for case-control association studies of heroin dependence. Previously, two single nucleotide polymorphisms (SNPs), OPRD1 921T > C and 80G > T, of the human delta opioid receptor gene were used in population-based studies of heroin dependence. One study in a German population found that OPRD1 921T > C was associated with heroin dependence. This finding, however, was not replicated in a different German sample. To test the hypothesis that OPRD1 or a closely linked gene is associated with heroin dependence, we used 5' nuclease assays to genotype both OPRD1 SNPs in 450 Chinese heroin dependent patients and 304 unaffected controls from the same population. In addition, five SNPs distributed in four other genes: ADH2, ALDH2, OPRM1, and DRD1, were used as genomic control loci to test the case and control populations for stratification bias. Genotype and allele frequencies at OPRD1 921T > C were not significantly different, and the OPRD1 80G was absent from both Chinese opioid dependence patients and controls. Based on the genotype and allele frequencies of the genomic control loci, there was no evidence for stratification bias capable of masking an association of OPRD1 to heroin dependence in this large and homogenous Chinese sample. Therefore, these data do not support an association between the OPRD1 gene and heroin dependence in the Chinese population.
Collapse
Affiliation(s)
- Ke Xu
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, NIH, Rockville, Maryland 20852, USA
| | | | | | | | | |
Collapse
|
759
|
Trujillo KA. The neurobiology of opiate tolerance, dependence and sensitization: mechanisms of NMDA receptor-dependent synaptic plasticity. Neurotox Res 2002; 4:373-91. [PMID: 12829426 DOI: 10.1080/10298420290023954] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Long-term administration of opiates leads to changes in the effects of these drugs, including tolerance, sensitization and physical dependence. There is, as yet, incomplete understanding of the neural mechanisms that underlie these phenomena. Tolerance, sensitization and physical dependence can be considered adaptive processes similar to other experience-dependent changes in the brain, such as learning and neural development. There is considerable evidence demonstrating that N-methyl-D-aspartate (NMDA) receptors and downstream signaling cascades may have an important role in different forms of experience-dependent changes in the brain and behavior. This review will explore evidence indicating that NMDA receptors and downstream messengers may be involved in opiate tolerance, sensitization and physical dependence. This evidence has been used to develop a cellular model of NMDA receptor/opiate interactions. According to this model, mu opioid receptor stimulation leads to a protein kinase C-mediated activation of NMDA receptors. Activation of NMDA receptors leads to influx of calcium and activation of calcium-dependent processes. These calcium-dependent processes have the ability to produce critical changes in opioid-responsive neurons, including inhibition of opioid receptor/second messenger coupling. This model is similar to cellular models of learning and neural development in which NMDA receptors have a central role. Together, the evidence suggests that the mechanisms that underlie changes in the brain and behavior produced by long-term opiate use may be similar to other central nervous system adaptations. The experimental findings and the resulting model may have implications for the treatment of pain and addiction.
Collapse
Affiliation(s)
- Keith A Trujillo
- Department of Psychology, California State University San Marcos, San Marcos, CA 92096-0001, USA.
| |
Collapse
|
760
|
Schinka JA, Town T, Abdullah L, Crawford FC, Ordorica PI, Francis E, Hughes P, Graves AB, Mortimer JA, Mullan M. A functional polymorphism within the mu-opioid receptor gene and risk for abuse of alcohol and other substances. Mol Psychiatry 2002; 7:224-8. [PMID: 11840318 DOI: 10.1038/sj.mp.4000951] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2000] [Revised: 02/16/2001] [Accepted: 02/23/2001] [Indexed: 11/09/2022]
Abstract
Genetic association studies investigating the role of the +118A allele of the human mu-opioid receptor gene in risk for alcohol dependency have produced inconsistent findings, possibly because of the failure to recognize sampling methodology difficulties inherent in association studies of polygenic disorders. We examined the frequency of the AA genotype and A allele in several groups of substance-dependent cases, unrestricted controls, and super controls screened for the use of alcohol and cigarettes. Our findings and analyses suggest that the OPRM1 +118 polymorphism is a general risk gene for substance dependence, but is not specific to a particular substance. The nature of the conferred risk is likely to be in use of multiple substances, but it is not yet determined if the risk could be expressed in severity of use of any particular substance. The contribution of the gene to risk for substance dependence is small, and is detected most easily in studies that use control samples that are screened for all forms of substance dependence.
Collapse
Affiliation(s)
- J A Schinka
- The Roskamp Institute, University of South Florida, Tampa, FL 33620, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
761
|
Chen WC, Wu HC, Hsu CD, Chen HY, Tsai FJ. p21 gene codon 31 polymorphism is associated with bladder cancer. Urol Oncol 2002; 7:63-6. [PMID: 12474524 DOI: 10.1016/s1078-1439(01)00152-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The function of p21 is related to cell apoptosis, progression and malignancies. It is thought that p21 is related to cancer formation but is not related to tumor grade. We aimed to investigative the polymorphism of p21 codon 31 as a candidate for the genetic marker of bladder cancer and its progression. The distribution was analyzed in 53 bladder cancer patients, 119 healthy controls in Taiwanese patients. Polymerase chain reaction based restriction analysis was used for the study of the association of p21 codon 31 polymorphism with bladder cancer. There was a significant difference in p21 codon 31 polymorphism between the control and the cancer patients (p < 0.01). The arginine form was prominent in the cancer patient (per copy of the A allele, odds ratio = 2.03, 95% confidence interval = 1.23-3.37). Furthermore, the distribution of this polymorphism was significantly different from non-invasive to invasive bladder cancer (p < 0.05). Serine heterozygote was more prominent in the invasive group with 25 to 1% respectively when compared with the non-invasive group. The polymorphism of p21 codon 31 is associated with bladder cancer. An individual possessing one allele of arginine form in p21 codon 31 has a higher risk of developing bladder cancer than the serine form. Although the mechanism is unclear, our results show p21 gene is associated with tumor grade.
Collapse
Affiliation(s)
- Wen-Chi Chen
- Department of Urology, China Medical College Hospital, School of Medicine, China Medical College, Taichung, Taiwan
| | | | | | | | | |
Collapse
|
762
|
Abstract
The sequencing of the human genome is only the first step. The next step is to determine the function of these genes and in particular, how alterations in specific genes lead to major human disorders. Many laboratories are now focusing on identifying and characterizing single nucleotide polymorphisms (SNPs), to determine which correlate in frequency with certain population groups who may be particularly susceptible to certain diseases. The mu opioid receptor (MOR), which mediates the clinically important analgesic effects of drugs like morphine as well as the euphoria sought by heroin abusers, exhibits several dozen polymorphisms. Several of these are associated with altered receptor function and individuals at risk for drug abuse.
Collapse
Affiliation(s)
- Nancy M Lee
- California Pacific Medical Center Research Institute, 2330 Clay Street, Stern Bldg., San Francisco, CA 94115, USA.
| | | |
Collapse
|
763
|
Abstract
Morphine-6-beta-glucuronide (M6G) is an opioid agonist that plays a role in the clinical effects of morphine. Although M6G probably crosses the blood-brain barrier with difficulty, during long term morphine administration it may reach sufficiently high CNS concentrations to exert clinically relevant opioid effects. As a consequence of its almost exclusive renal elimination, M6G may accumulate in the body of patients with impaired renal function and cause severe opioid adverse effects with insidious onset and long persistence. Its profile of receptor affinities, however, gives reason to speculate that M6G may exhibit analgesic effects while causing fewer adverse effects than morphine. This is supported by reports of the good tolerability of intrathecal and intravenous injections of M6G in humans with intact renal function. M6G may thus be contemplated as an analgesic for short term postoperative analgesia, especially for intrathecal analgesic therapy. In addition, its possibly higher potency than morphine makes M6G a candidate opioid for local or peripheral analgesic therapy. However, current knowledge is too incomplete to finally judge the clinical usefulness of M6G. The next topics for clinical research on M6G should include: (i) a comparison of the potencies of M6G and morphine to cause wanted and unwanted clinical effects; (ii) development of a predictive population pharmacokinetic-pharmacodynamic model of M6G with calculation of the transfer half-life between plasma and effect site; and (iii) identification of cofactors influencing the action of M6G that can serve as predictors for the clinical outcome of morphine/M6G therapy in an individual including the pharmacogenetics of M6G.
Collapse
Affiliation(s)
- J Lötsch
- Johann-Wolfgang-Goethe-Universität, Frankfurt am Main, Germany.
| | | |
Collapse
|
764
|
Lötsch J, Skarke C, Grösch S, Darimont J, Schmidt H, Geisslinger G. The polymorphism A118G of the human mu-opioid receptor gene decreases the pupil constrictory effect of morphine-6-glucuronide but not that of morphine. PHARMACOGENETICS 2002; 12:3-9. [PMID: 11773859 DOI: 10.1097/00008571-200201000-00002] [Citation(s) in RCA: 154] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Large individual differences in the clinical response to morphine therapy have been known for a long time by clinicians. The recent advances in genomic research encourage the search for pharmacogenetic causes of that variability. As a measure of central opioid effects, pupil diameters were assessed every 20 min for 18 h after administration of morphine or its active metabolite morphine-6-glucuronide (M6G) in a two-way crossover study. The opioid effects were compared between six subjects with a single-nucleotide polymorphism (SNP) A118G in the mu-opioid receptor gene (five heterozygous, one homozygous) and six control subjects. Non-parametric pharmacokinetic-pharmacodynamic modelling was employed to identify the influence of the A118G SNP on the concentration-response relationship of M6G and morphine, which was described by a sigmoid Emax model. As a measure of potency, the EC50 of the pupil constrictory effects of M6G was 714 +/- 197 nmol/l in wild-type and 1475 +/- 424 nmol/l in heterozygous carriers of the A118G SNP. In the homozygous carrier of the SNP, it had an EC50 of 3140 nmol/l. In addition, the dose-response relationship was flatter in the A118G carriers than in control subjects (shape factor of the sigmoid Emax model: gamma = 3.3 +/- 1.2, 1.7 +/- 0.5 and 1.6 for wild-type, heterozygous and the homozygous A118G carriers, respectively). In contrast, the concentration-response relationship of morphine was not affected by this specific SNP. The A118G SNP in the mu-receptor gene significantly reduces the potency of M6G in humans.
Collapse
Affiliation(s)
- Jörn Lötsch
- Pharmazentrum Frankfurt, Johann Wolfgang Goethe-University, Frankfurt, Germany.
| | | | | | | | | | | |
Collapse
|
765
|
Höllt V. A polymorphism (A118G) in the mu-opioid receptor gene affects the response to morphine-6-glucuronide in humans. PHARMACOGENETICS 2002; 12:1-2. [PMID: 11773858 DOI: 10.1097/00008571-200201000-00001] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Volker Höllt
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
766
|
Grösch S, Niederberger E, Lötsch J, Skarke C, Geisslinger G. A rapid screening method for a single nucleotide polymorphism (SNP) in the human MOR gene. Br J Clin Pharmacol 2001. [PMID: 11736886 PMCID: PMC2014560 DOI: 10.1046/j.1365-2125.2001.01504.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2023] Open
Abstract
AIMS Genetic association studies have suggested that the single nucleotide polymorphism (SNP) at position 118 of the human mu-opioid receptor (MOR) gene could be a potential risk factor for drug treatment variability in patients. Therefore, we wanted to develop a fast and reliable detection method for this SNP which is applicable in a clinical setting. METHODS To detect the polymorphism at position A118-->G in the human MOR gene we used the fluorescence resonance energy transfer (FRET)-PCR technique with subsequent melting curve analysis. RESULTS The polymorphism at position A118-->G in the human MOR gene could be clearly discriminated with melting peak temperatures of 69.8 degrees C and 63.8 degrees C, corresponding to the wild type and mutated MOR allele, respectively. The results from FRET-PCR were validated by sequencing and restriction-fragment length polymorphism (RFLP). Screening of blood samples from 100 subjects showed an allelic distribution for the human MOR alleles of 79% (homozygous wild type), 20% (heterozygous) and 0.9% (homozygous mutated). CONCLUSIONS The FRET-PCR protocol for detection of the human MOR gene polymorphism at position 118 offers a rapid and reliable method which could be used for population screening of this and other genes.
Collapse
Affiliation(s)
- S Grösch
- pharmazentrum frankfurt, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt, Theodor Stern Kai7, 60590 Frankfurt/Main, Germany
| | | | | | | | | |
Collapse
|
767
|
Grösch S, Niederberger E, Lötsch J, Skarke C, Geisslinger G. A rapid screening method for a single nucleotide polymorphism (SNP) in the human MOR gene. Br J Clin Pharmacol 2001; 52:711-4. [PMID: 11736886 PMCID: PMC2014560 DOI: 10.1046/j.0306-5251.2001.01504.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AIMS Genetic association studies have suggested that the single nucleotide polymorphism (SNP) at position 118 of the human mu-opioid receptor (MOR) gene could be a potential risk factor for drug treatment variability in patients. Therefore, we wanted to develop a fast and reliable detection method for this SNP which is applicable in a clinical setting. METHODS To detect the polymorphism at position A118-->G in the human MOR gene we used the fluorescence resonance energy transfer (FRET)-PCR technique with subsequent melting curve analysis. RESULTS The polymorphism at position A118-->G in the human MOR gene could be clearly discriminated with melting peak temperatures of 69.8 degrees C and 63.8 degrees C, corresponding to the wild type and mutated MOR allele, respectively. The results from FRET-PCR were validated by sequencing and restriction-fragment length polymorphism (RFLP). Screening of blood samples from 100 subjects showed an allelic distribution for the human MOR alleles of 79% (homozygous wild type), 20% (heterozygous) and 0.9% (homozygous mutated). CONCLUSIONS The FRET-PCR protocol for detection of the human MOR gene polymorphism at position 118 offers a rapid and reliable method which could be used for population screening of this and other genes.
Collapse
Affiliation(s)
- S Grösch
- pharmazentrum frankfurt, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt, Theodor Stern Kai7, 60590 Frankfurt/Main, Germany
| | | | | | | | | |
Collapse
|
768
|
Ohmori O, Shinkai T, Hori H, Kojima H, Nakamura J. Polymorphisms of mu and delta opioid receptor genes and tardive dyskinesia in patients with schizophrenia. Schizophr Res 2001; 52:137-8. [PMID: 11595401 DOI: 10.1016/s0920-9964(00)00188-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
769
|
Wang D, Quillan JM, Winans K, Lucas JL, Sadée W. Single nucleotide polymorphisms in the human mu opioid receptor gene alter basal G protein coupling and calmodulin binding. J Biol Chem 2001; 276:34624-30. [PMID: 11457836 DOI: 10.1074/jbc.m104083200] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The mu opioid receptor (MOR) plays a central role in mediating acute and chronic effects of narcotic drugs. Three rare single nucleotide polymorphisms in the hMOR gene have been identified that cause amino acid substitutions in the third intracellular (i3) loop of MOR (R260H, R265H, and S268P). Genotyping 252 individuals of the Coriell collection identified one allele encoding the R265H-MOR variant and a new variant encoding D274N-MOR. Variants R260H-, R265H-, and S268P-MOR were constructed and transfected into HEK293 cells. Morphine stimulated G protein coupling of the three receptor variants to a maximal level approaching that of wild type MOR. In contrast, spontaneous, agonist-independent (basal) MOR signaling, proposed to play a role in opioid tolerance and dependence, was significantly reduced for R260H- and R265H-MOR. Moreover, domains within the i3 loop of MOR have been shown to interact with both G proteins and calmodulin (CaM). CaM binding was deficient for variants R265H- and S268P-MOR, suggesting that domains for G protein coupling and CaM binding overlap partially. Morphine pretreatment significantly enhanced basal G protein coupling of wild type MOR, which is thought to result from release of CaM. In contrast basal G protein coupling activity of the three variants was unaffected by morphine pretreatment consistent with diminished CaM regulation, low basal activity, or both. In conclusion, each of the three single nucleotide polymorphisms mapping to the i3 loop of MOR caused substantial changes in basal G protein coupling, CaM binding, or both. Carriers of the mutant alleles might display altered responses to narcotic analgesics.
Collapse
Affiliation(s)
- D Wang
- Department of Biopharmaceutical Sciences and Pharmaceutical Chemistry, University of California, San Francisco, California 94143-0446, USA
| | | | | | | | | |
Collapse
|
770
|
Abstract
People with a genetic predisposition for substance abuse have defects in genes for the opioid peptides and receptors. A high number of polymorphisms have been detected in the mu-opioid receptor, some of which result in pharmacological alterations. The opioid peptide proopiomelanocortin proved extraordinarily rich in mutations that often lead to severe phenotypical consequences. Prodynorphin displays a polymorphic regulation of transcription. Variants of the mu- and the delta-opioid receptor showed positive associations with opiate and/or alcohol addiction in some studies. However, these associations were weak, indicating a small contribution of the opioid system to these disorders.
Collapse
MESH Headings
- Alleles
- Enkephalins/genetics
- Enkephalins/pharmacology
- Genetic Predisposition to Disease
- Humans
- Polymorphism, Genetic
- Protein Precursors/genetics
- Protein Precursors/pharmacology
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/physiology
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/physiology
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/physiology
- Substance-Related Disorders/genetics
- beta-Endorphin/genetics
- beta-Endorphin/pharmacology
Collapse
Affiliation(s)
- P Mayer
- Institute for Pharmacology and Toxicology, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | | |
Collapse
|
771
|
Cigler T, LaForge KS, McHugh PF, Kapadia SU, Leal SM, Kreek MJ. Novel and previously reported single-nucleotide polymorphisms in the human 5-HT(1B) receptor gene: no association with cocaine or alcohol abuse or dependence. AMERICAN JOURNAL OF MEDICAL GENETICS 2001; 105:489-97. [PMID: 11496363 PMCID: PMC6148750 DOI: 10.1002/ajmg.1473] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Evidence from animal self-administration and human genetics studies suggests that the serotonin(1B) (5-HT(1B)) receptor may be involved in modulating responses to cocaine or alcohol. We hypothesize that polymorphisms, including single-nucleotide polymorphisms (SNPs), in the human 5-HT(1B) receptor gene, may be associated with individual differences in vulnerability to cocaine or alcohol abuse or dependence. A total of 210 subjects were studied, including individuals with a primary diagnosis (DSM-IV criteria) of cocaine abuse or dependence, alcohol abuse or dependence, and controls with no history of previous or current illicit drug or alcohol abuse or dependence. Genomic DNA samples were isolated from each individual. For 157 of the subjects, polymerase chain reaction (PCR) was used to amplify the entire coding region of the 5-HT(1B) receptor gene as well as parts of the 5' and 3' untranslated regions. PCR products were sequenced in forward and reverse directions on an automated sequencer. Amplified DNA from an additional 53 subjects was sequenced in the 5' untranslated region to gain additional data on the frequency of one identified SNP. Seven polymorphisms were identified: one novel SNP in the 5' untranslated region (UTR) of the gene (A-161T); one SNP not reported in any published scientific communication (but found to be recorded in GenBank) in the 3' UTR (A1180G); two novel dinucleotide deletions at positions - 184/- 183 and - 182/- 181; and three previously identified SNPs (T-261G, C129T, G861C). Data were stratified by ethnicity and pooled Relative Risk was calculated for combined alcohol abuse and dependence cases and controls, and also for combined cocaine abuse and dependence cases and controls. No significant differences between cases and controls were found.
Collapse
Affiliation(s)
- Tessa Cigler
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, New York
| | - K. Steven LaForge
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, New York
| | - Pauline F. McHugh
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, New York
| | - Sagar U. Kapadia
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, New York
| | - Suzanne M. Leal
- Laboratory of Statistical Genetics, Rockefeller University, New York, New York
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, New York
| |
Collapse
|
772
|
Clinical pharmacology of morphine and morphine-6-glucuronide. A PK/PD modeling approach. ACTA ACUST UNITED AC 2001. [DOI: 10.1016/s0531-5131(01)00284-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
773
|
Sadee W, Hoeg E, Lucas J, Wang D. Genetic variations in human G protein-coupled receptors: implications for drug therapy. AAPS PHARMSCI 2001; 3:E22. [PMID: 11741273 PMCID: PMC2751017 DOI: 10.1208/ps030322] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Numerous genes encode G protein-coupled receptors (GPCRs)-a main molecular target for drug therapy. Estimates indicate that the human genome contains approximately 600 GPCR genes. This article addresses therapeutic implications of sequence variations in GPCR genes. A number of inactivating and activating receptor mutations have been shown to cause a variety of (mostly rare) genetic disorders. However, pharmacogenetic and pharmacogenomic studies on GPCRs are scarce, and therapeutic relevance of variant receptor alleles often remains unclear. Confounding factors in assessing the therapeutic relevance of variant GPCR alleles include 1) interaction of a single drug with multiple closely related receptors, 2) poorly defined binding pockets that can accommodate drug ligands in different orientations or at alternative receptor domains, 3) possibility of multiple receptor conformations with distinct functions, and 4) multiple signaling pathways engaged by a single receptor. For example, antischizophrenic drugs bind to numerous receptors, several of which might be relevant to therapeutic outcome. Without knowing accurately what role a given receptor subtype plays in clinical outcome and how a sequence variation affects drug-induced signal transduction, we cannot predict the therapeutic relevance of a receptor variant. Genome-wide association studies with single nucleotide polymorphisms could identify critical target receptors for disease susceptibility and drug efficacy or toxicity.
Collapse
Affiliation(s)
- W Sadee
- Department of Biopharmaceutical Sciences, University of California San Francisco, San Francisco CA 94143-0446, USA.
| | | | | | | |
Collapse
|
774
|
Compton P, Charuvastra VC, Ling W. Pain intolerance in opioid-maintained former opiate addicts: effect of long-acting maintenance agent. Drug Alcohol Depend 2001; 63:139-46. [PMID: 11376918 DOI: 10.1016/s0376-8716(00)00200-3] [Citation(s) in RCA: 213] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Patients on methadone maintenance therapy are relatively intolerant of pain, a finding hypothesized to reflect a hyperalgesic state induced by chronic opioid administration. To explore if the intrinsic activity of the opioid maintenance agent might affect expression of hyperalgesia in this population, withdrawal latency for cold-pressor (CP) pain was compared between small groups of methadone-maintained (n = 18), buprenorphine-maintained (n = 18), and matched control (n = 18) subjects. The opioid-maintained groups had equal and significantly shorter withdrawal latencies than controls, however it is possible that high rates of continued illicit opioid use precluded finding differences between methadone and buprenorphine groups. Differential effects of maintenance agent were found for the few subjects without illicit opioid use, such that withdrawal latencies for methadone-maintained (n = 5) were less than for buprenorphine-maintained (n = 7) which were less than controls (n = 18). Diminished pain tolerance in patients receiving opioid maintenance treatment has significant clinical implications. More research is needed to determine if buprenorphine offers advantages over methadone in this regard.
Collapse
Affiliation(s)
- P Compton
- School of Nursing, University of California at Los Angeles (UCLA), Factor Building 4-246, Box 956918, Los Angeles, CA 90095-6918, USA.
| | | | | |
Collapse
|
775
|
Chavkin C, McLaughlin JP, Celver JP. Regulation of opioid receptor function by chronic agonist exposure: constitutive activity and desensitization. Mol Pharmacol 2001; 60:20-5. [PMID: 11408596 DOI: 10.1124/mol.60.1.20] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- C Chavkin
- Department of Pharmacology, University of Washington, Seattle, Washington 98195-7280, USA.
| | | | | |
Collapse
|
776
|
Rana BK, Shiina T, Insel PA. Genetic variations and polymorphisms of G protein-coupled receptors: functional and therapeutic implications. Annu Rev Pharmacol Toxicol 2001; 41:593-624. [PMID: 11264470 DOI: 10.1146/annurev.pharmtox.41.1.593] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
G protein-coupled receptors (GPCRs) represent a major class of proteins in the genome of many species, including humans. In addition to the mapping of a number of human disorders to regions of the genome containing GPCRs, a growing body of literature has documented frequently occurring variations (i.e. polymorphisms) in GPCR loci. In this article, we use a domain-based approach to systematically examine examples of genetic variation in the coding and noncoding regions of GPCR loci. Data to date indicate that residues in GPCRs are involved in ligand binding and coupling to G proteins and that regulation can be altered by polymorphisms. Studies of GPCR polymorphisms have also uncovered the functional importance of residues not previously implicated from other approaches that are involved in the function of GPCRs. We predict that studies of GPCR polymorphisms will have a significant impact on medicine and pharmacology, in particular, by providing new means to subclassify patients in terms of both diagnosis and treatment.
Collapse
Affiliation(s)
- B K Rana
- Department of Pharmacology, University of California at San Diego, La Jolla, California 92093-0636, USA.
| | | | | |
Collapse
|
777
|
The untranslated region of (mu)-opioid receptor mRNA contributes to reduced opioid sensitivity in CXBK mice. J Neurosci 2001. [PMID: 11160404 DOI: 10.1523/jneurosci.21-04-01334.2001] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
It is well known that there are individual differences in a sensitivity to analgesics. Several lines of evidence have suggested that the level of opioid-induced analgesia is dependent on the level of expression of the mu-opioid receptor (mu-OR). However, the molecular mechanisms underlying the diversity of the level of the opioid receptor and the opioid sensitivity among individuals remain to be elucidated. In the present study, we analyzed the opioid-receptor genes of CXBK recombinant-inbred mice, which show reduced sensitivity to opioids. Northern blotting, nucleotide sequencing, and in situ hybridization histochemical analyses demonstrated that CXBK mice possessed mu-OR mRNA with a normal coding region but an abnormally long untranslated region (UTR). In addition, the mu-OR mRNA level in CXBK mice was less than in the control mice. Next, we produced littermate mice that had inherited two copies of the wild-type mu-OR gene, had inherited two copies of the CXBK mu-OR gene, and had inherited both copies of the mu-OR genes. In these mice, inheritance of the CXBK mu-OR gene was well correlated with less mu-OR mRNA and reduced opioid effects on nociception and locomotor activity. We conclude that the CXBK mu-OR gene is responsible for the CXBK phenotypes. Because UTR differences are known to affect the level of the corresponding mRNA and protein and because UTRs are more divergent among individuals than coding regions, the present findings suggest that opioid sensitivity may vary, depending on different mu-OR levels attributable to divergent UTR of mu-OR mRNA.
Collapse
|
778
|
Abstract
Pharmacogenomics deals with the interactions of individual genetic constitution with drug therapy. It has potentially far reaching consequences for drug development and future treatment strategies, but also for clinical in vitro diagnostics. With increasing knowledge about interactions between genes and drug treatment, there will be an equally increasing demand for rapid and reliable diagnostic tests prior to the institution of therapy. In fact, it is very likely that pharmacogenetic tests will make up a significant proportion of total molecular biology testing in the coming years. Therefore, this review focuses on the implications of pharmacogenomics on the clinical laboratory.
Collapse
Affiliation(s)
- G Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, D-93042 Regensburg, Germany.
| | | | | |
Collapse
|
779
|
Rommelspacher H, Smolka M, Schmidt LG, Samochowiec J, Hoehe MR. Genetic analysis of the mu-opioid receptor in alcohol-dependent individuals. Alcohol 2001; 24:129-35. [PMID: 11522434 DOI: 10.1016/s0741-8329(01)00139-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
On the basis of various study results, it is suggested that the ethanol-induced activation of the endogenous opioid system may play an important role in mediating the reinforcing effects of ethanol. The mesolimbic dopamine reward system is activated by both ethanol and opioids, and genetic differences in the sensitivity of the endogenous opioid system to alcohol may be an important factor determining the risk for the development of excessive alcohol consumption. Thus, variants of the mu-opioid receptor (muOR) gene may confer vulnerability to alcohol dependence. Five exon 1 variants of the muOR were investigated in 327 alcohol-dependent and 340 healthy control subjects. The Val6 variant of the +17C/T polymorphism and the Asp40 variant of the +118A/G polymorphism showed a trend to an increased allele frequency in alcohol-dependent subjects. The latter polymorphism was investigated in more detail. The dopamine receptor agonist apomorphine causes an increase in growth hormone (GH) levels in the blood by stimulating the release of growth hormone-releasing hormone. beta-endorphin also activates this regulatory circuit. We found a blunted response in intoxicated alcohol-dependent subjects, but no difference in GH response between the groups of alcohol-dependent subjects with and without the variant Asp allele. However, alcohol-dependent subjects with the Asp allele showed a significantly higher GH response at day 7 after alcohol withdrawal and a tendency to lower novelty seeking. These results suggest to us that there is reduced dopaminergic neuronal activity in alcohol-dependent subjects with the muOR Asp40 allele, along with a compensating increase in dopamine receptor activity. The difference between the two groups of alcohol-dependent subjects can be demonstrated only under certain conditions such as alcohol withdrawal, which necessitates the adaptation of the neurones to a new homeostasis.
Collapse
Affiliation(s)
- H Rommelspacher
- Department of Clinical Neurobiology, Free University, Ulmenallee 32, 14050 Berlin, Germany.
| | | | | | | | | |
Collapse
|
780
|
Szeto CY, Tang NL, Lee DT, Stadlin A. Association between mu opioid receptor gene polymorphisms and Chinese heroin addicts. Neuroreport 2001; 12:1103-6. [PMID: 11338173 DOI: 10.1097/00001756-200105080-00011] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Mu opioid receptor (MOR) has been shown to be associated with alcoholism and opioid dependence. The present study examined the involvement of a polymorphism in A118G in exon 1 and C1031G in intron 2 of the MOR gene in 200 Chinese heroin-dependent and 97 control subjects. Results showed a significant association for both A118G and C1031G polymorphisms and opioid dependence. The G allele is more common in the heroin-dependent group (39.5% and 30.8% for A118G and C1031G polymorphisms, respectively) when compared to the controls (29.4% and 21.1% for A118G and C1031G polymorphisms, respectively). This study suggests that the variant G allele of both A118G and C1031G polymorphisms may contribute to the vulnerability to heroin dependence.
Collapse
Affiliation(s)
- C Y Szeto
- Department of Anatomy, Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | | | | | | |
Collapse
|
781
|
Mann K, Agartz I, Harper C, Shoaf S, Rawlings RR, Momenan R, Hommer DW, Pfefferbaum A, Sullivan EV, Anton RF, Drobes DJ, George MS, Bares R, Machulla HJ, Mundle G, Reimold M, Heinz A. Neuroimaging in Alcoholism: Ethanol and Brain Damage. Alcohol Clin Exp Res 2001. [DOI: 10.1111/j.1530-0277.2001.tb02383.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
782
|
Gant TM, Riba P, Lee NM. Morphine tolerance in mice is independent of polymorphisms in opioid receptor sequences. Brain Res Bull 2001; 55:59-63. [PMID: 11427338 DOI: 10.1016/s0361-9230(01)00497-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Pharmacogenomics links individual drug response variation to genetic differences, such as single nucleotide polymorphisms (SNPs). In particular, pharmacogenomics will allow clinicians to use genetic diagnostics to predict the response of a patient to a drug. We investigated whether SNPs in opioid receptors correlated with the development of morphine tolerance in mouse strains that showed either high or low tolerance to morphine. Sequencing identified five silent SNPs in the delta opioid receptor that varied from the published sequence in some strains, but which were found in both high and low tolerance strains. The mu and kappa opioid receptor sequences had no SNPs. Taken together, these data definitively demonstrate that morphine tolerance development in mice is independent of opioid receptor sequence.
Collapse
Affiliation(s)
- T M Gant
- Geraldine Brush Cancer Research Institute, California Pacific Medical Center Research Institute, San Francisco, CA 94115, USA
| | | | | |
Collapse
|
783
|
Ikeda K, Kobayashi T, Ichikawa T, Kumanishi T, Niki H, Yano R. The untranslated region of (mu)-opioid receptor mRNA contributes to reduced opioid sensitivity in CXBK mice. J Neurosci 2001; 21:1334-9. [PMID: 11160404 PMCID: PMC6762225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
It is well known that there are individual differences in a sensitivity to analgesics. Several lines of evidence have suggested that the level of opioid-induced analgesia is dependent on the level of expression of the mu-opioid receptor (mu-OR). However, the molecular mechanisms underlying the diversity of the level of the opioid receptor and the opioid sensitivity among individuals remain to be elucidated. In the present study, we analyzed the opioid-receptor genes of CXBK recombinant-inbred mice, which show reduced sensitivity to opioids. Northern blotting, nucleotide sequencing, and in situ hybridization histochemical analyses demonstrated that CXBK mice possessed mu-OR mRNA with a normal coding region but an abnormally long untranslated region (UTR). In addition, the mu-OR mRNA level in CXBK mice was less than in the control mice. Next, we produced littermate mice that had inherited two copies of the wild-type mu-OR gene, had inherited two copies of the CXBK mu-OR gene, and had inherited both copies of the mu-OR genes. In these mice, inheritance of the CXBK mu-OR gene was well correlated with less mu-OR mRNA and reduced opioid effects on nociception and locomotor activity. We conclude that the CXBK mu-OR gene is responsible for the CXBK phenotypes. Because UTR differences are known to affect the level of the corresponding mRNA and protein and because UTRs are more divergent among individuals than coding regions, the present findings suggest that opioid sensitivity may vary, depending on different mu-OR levels attributable to divergent UTR of mu-OR mRNA.
Collapse
MESH Headings
- Animals
- Brain/metabolism
- DNA Mutational Analysis
- DNA, Complementary/analysis
- DNA, Complementary/genetics
- Drug Resistance/genetics
- Gene Dosage
- Heterozygote
- Homozygote
- In Situ Hybridization
- Male
- Mice
- Mice, Inbred Strains
- Mice, Transgenic
- Molecular Weight
- Morphine/pharmacology
- Motor Activity/drug effects
- Motor Activity/genetics
- Pain Measurement/drug effects
- Point Mutation
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Untranslated/genetics
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, mu/genetics
Collapse
Affiliation(s)
- K Ikeda
- Laboratories for Neurobiology of Emotion and Cellular Information Processing, Brain Science Institute, RIKEN, Wako, Saitama 351-0198, Japan.
| | | | | | | | | | | |
Collapse
|
784
|
Befort K, Filliol D, Decaillot FM, Gaveriaux-Ruff C, Hoehe MR, Kieffer BL. A single nucleotide polymorphic mutation in the human mu-opioid receptor severely impairs receptor signaling. J Biol Chem 2001; 276:3130-7. [PMID: 11067846 DOI: 10.1074/jbc.m006352200] [Citation(s) in RCA: 189] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Large scale sequencing of the human mu-opioid receptor (hMOR) gene has revealed polymorphic mutations that occur within the coding region. We have investigated whether the mutations N40D in the extracellular N-terminal region, N152D in the third transmembrane domain, and R265H and S268P in the third intracellular loop alter functional properties of the receptor expressed in mammalian cells. The N152D receptor was produced at low densities. Binding affinities of structurally diverse opioids (morphine, diprenorphine, DAMGO and CTOP) and the main endogenous opioid peptides (beta-endorphin, [Met]enkephalin, and dynorphin A) were not markedly changed in mutant receptors (<3-fold). Receptor signaling was strongly impaired in the S268P mutant, with a reduction of efficacy and potency of several agonists (DAMGO, beta-endorphin, and morphine) in two distinct functional assays. Signaling at N40D and R265H mutants was highly similar to wild type, and none of the mutations induced detectable constitutive activity. DAMGO-induced down-regulation of receptor-binding sites, following 20 h of treatment, was identical in wild-type and mutant receptors. Our data show that natural sequence variations in hMOR gene have little influence on ligand binding or receptor down-regulation but could otherwise modify receptor density and signaling. Importantly, the S268P mutation represents a loss-of-function mutation for the human mu-opioid receptor, which may have an incidence on opioid-regulated behaviors or drug addiction in vivo.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Asparagine/genetics
- Aspartic Acid/genetics
- COS Cells
- Cells, Cultured
- Cyclic AMP/metabolism
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Guanosine 5'-O-(3-Thiotriphosphate)/metabolism
- Humans
- Mutagenesis, Site-Directed
- Narcotics/pharmacology
- Polymorphism, Single Nucleotide
- Proline/genetics
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Serine/genetics
- Signal Transduction/physiology
- Sulfur Radioisotopes
Collapse
Affiliation(s)
- K Befort
- Laboratoire des Récepteurs et Protéines Membranaires, UPR CNRS 9050, ESBS, Parc d'Innovation, 67400 Illkirch, France
| | | | | | | | | | | |
Collapse
|
785
|
Fagerlund TH, Braaten O. No pain relief from codeine...? An introduction to pharmacogenomics. Acta Anaesthesiol Scand 2001. [DOI: 10.1034/j.1399-6576.2001.450203.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
786
|
Franke P, Wang T, N�then MM, Knapp M, Neidt H, Albrecht S, Jahnes E, Propping P, Maier W. Nonreplication of association between ?-opioid-receptor gene (OPRM1) A118G polymorphism and substance dependence. ACTA ACUST UNITED AC 2001. [DOI: 10.1002/1096-8628(20010108)105:1<114::aid-ajmg1074>3.0.co;2-l] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
787
|
Abstract
The risk of initiating and maintaining the use of opiates up to the point of abuse and dependence is to a large degree genetically transmitted and is separate from genetic risk factors for addiction to other drugs of abuse. Pharmacogenetic studies have so far focused on obvious candidate genes that are expected to be involved either in the pharmacokinetics or in the pharmacodynamics of opioids in the mesolimbic reward system of the brain. The few findings of a positive allelic association rarely withstand replication in independent case-control or less stratification-prone family-based association samples. A pharmacogenomic approach in the best sense of the word, however, involves an unbiased, genome-wide, parallel search for risk genes and gene expression patterns. So far, only quantitative trait loci mapping studies of inbred rodent strains and differential expression studies using high-density DNA microarrays fulfill these requirements. The present state of pharmacogenomic and pharmacogenetic studies in animals and humans with respect to opiate addiction is reviewed in this paper.
Collapse
Affiliation(s)
- D Lichtermann
- Department of Psychiatry, University of Bonn, Sigmund-Freud-Str. 25, D-53105, Bonn, Germany.
| | | | | | | |
Collapse
|
788
|
Town T, Schinka J, Tan J, Mullan M. The opioid receptor system and alcoholism: a genetic perspective. Eur J Pharmacol 2000; 410:243-248. [PMID: 11134673 DOI: 10.1016/s0014-2999(00)00818-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Over the past decade, mounting evidence has implicated the endogenous opioid receptor system as a central player in the etiology of alcohol drinking behavior in animals and alcoholism in humans. Much of this work is a product of a pharmacological approach, where differences in opioid receptor pharmacology have been found to predict drinking behavior in animal models of alcoholism, including rats and mice selectively bred for alcohol preference and avoidance. This review considers the opioid receptor system and alcoholism from a genetic standpoint, and discusses investigation into opioid receptor pharmacology in animal models of alcoholism as work that paved the way for the more recent molecular genetic studies implicating the delta-, and particularly, the mu opioid receptors as genetically linked to alcoholism-associated phenotypes in animal models of the disease. These genetic studies are set within the broader context of the candidate gene approach for alcoholism, where opioid receptor genes are taken to be partial, rather than complete, risk factors for alcoholism. Building upon these findings, the recent genetic association between alcoholism and the mu opioid receptor gene in humans is discussed. Finally, the translation of such genetic association studies between opioid receptor genes and alcoholism to a pharmacogenetic approach, allowing for the evaluation of putative relationships between genotype and pharmacological response profiles, is suggested to address the etiological question of what the molecular mechanism is underlying opioid receptor genetic risk for alcoholism phenotypes.
Collapse
Affiliation(s)
- T Town
- The Roskamp Institute, 3515 E. Fletcher Ave., Tampa, FL 33613, USA.
| | | | | | | |
Collapse
|
789
|
LaForge KS, Yuferov V, Kreek MJ. Opioid receptor and peptide gene polymorphisms: potential implications for addictions. Eur J Pharmacol 2000; 410:249-268. [PMID: 11134674 DOI: 10.1016/s0014-2999(00)00819-0] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Addictions to drugs of abuse and alcohol have been shown by studies of genetic epidemiology to have both a heritable and an environmental basis, with these factors influencing addiction to different substances to a different extent. In the search for specific alleles of specific genes that may contribute to the development of the addictions, many researchers have focused on the endogenous opioid system, which mediates a diverse array of neurological, physiological, and behavioral functions. The endogenous opioid system is also centrally important in mediating the effects of drugs of abuse and alcohol. Polymorphisms, including single nucleotide polymorphisms, have been identified in genes of the endogenous opioid receptors and peptides. A number of recent genetic association studies and a few studies of potential function provide clues as to which genes and which alleles may have implications for human physiology and pathophysiology, including the addictions.
Collapse
Affiliation(s)
- K S LaForge
- Laboratory Biology of Addictive Diseases, The Rockefeller University, Box 171, 1230 York Avenue, New York, NY 10021, USA.
| | | | | |
Collapse
|
790
|
LaForge KS, Shick V, Spangler R, Proudnikov D, Yuferov V, Lysov Y, Mirzabekov A, Kreek MJ. Detection of single nucleotide polymorphisms of the human mu opioid receptor gene by hybridization or single nucleotide extension on custom oligonucleotide gelpad microchips: potential in studies of addiction. AMERICAN JOURNAL OF MEDICAL GENETICS 2000; 96:604-15. [PMID: 11054767 DOI: 10.1002/1096-8628(20001009)96:5<604::aid-ajmg5>3.0.co;2-f] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The human mu opioid receptor (MOR) plays a central role in mediating the effects of opioids, both endogenous and exogenous. Epidemiological studies have shown that addiction in general, and especially opiate addiction, has a heritable component. Clinical and laboratory studies suggest that the MOR gene may contribute to the heritable component of vulnerability to develop opiate addiction. Naturally occurring single nucleotide polymorphisms (SNPs) have been identified in the MOR gene by conventional methods. Two coding region SNPs, the A118G and C17T substitutions, occur at high allelic frequencies (10.5% and 6.6%, respectively, in our previous studies). These common SNPs cause amino acid changes in the receptor, and may have implications for differences in individual responses to opioids, as well as decreased or increased vulnerability to opiate addiction. The A118G substitution encodes a variant receptor with binding and signal transduction differences in response to beta-endorphin in cellular assays. Recent innovations in microchip technology offer new potential methods for SNP detection. We report here on the development of two separate approaches using custom oligonucleotide gelpad microarrays for detection of these two common SNPs of the MOR gene in human DNA samples. First, PCR-amplified genomic DNA samples were used to produce target sequences, which were labeled with fluorescent dye and hybridized to custom microchips. Oligonucleotides on these reusable microchips were designed to query nucleotide substitutions at positions 17 and 118 of the MOR gene. Thirty-six human DNA samples were assayed both on these custom microchips and by conventional automated gel sequencing, with highly concordant identification of both heterozygous and homozygous substitutions. A second approach was developed for the C17T SNP utilizing single nucleotide extension on custom microchips. These custom gelpad microchips have potential for the rapid and inexpensive detection of specific SNPs for genetic and genomic studies.
Collapse
Affiliation(s)
- K S LaForge
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York 10021, USA
| | | | | | | | | | | | | | | |
Collapse
|
791
|
Abstract
Providing pain management for known opioid abusers is a challenging clinical task, in part because little is known about their pain experience and analgesic requirements. This study was designed to describe pain tolerance and analgesic response in a sample of opioid addicts stabilized in methadone-maintenance (MM) treatment (n = 60) in comparison to matched nondependent control subjects (n = 60). By using a placebo-controlled, two-way factorial design, tolerance to cold-pressor (CP) pain was examined, both before and after oral administration of therapeutic doses of common opioid (hydromorphone 2 mg) and nonsteroidal anti-inflammatory (ketorolac 10 mg) analgesic agents. Results showed that MM individuals were significantly less tolerant of CP pain than control subjects, replicating previous work. Analgesic effects were significant neither for medication nor group. These data indicate that MM opioid abusers represent a pain-intolerant subset of clinical patients. Their complaints of pain should be evaluated seriously and managed aggressively.
Collapse
Affiliation(s)
- P Compton
- School of Nursing, University of California at Los Angeles, Los Angeles, CA 90095-6918, USA
| | | | | | | |
Collapse
|
792
|
Abstract
Cloning of multiple opioid receptors has presented opportunities to investigate the mechanisms of multiple opioid receptor signaling and the regulation of these signals. The subsequent identification of receptor gene structures has also provided opportunities to study the regulation of receptor gene expression and to manipulate the concentration of the gene products in vivo. Thus, in the current review, we examine recent advances in the delineation basis for the multiple opioid receptor signaling, and their regulation at multiple levels. We discuss the use of receptor knockout animals to investigate the function and the pharmacology of these multiple opioid receptors. The reasons and basis for the multiple opioid receptor are addressed.
Collapse
Affiliation(s)
- P Y Law
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis 55455, USA.
| | | | | |
Collapse
|
793
|
Kreek MJ. Methadone-related opioid agonist pharmacotherapy for heroin addiction. History, recent molecular and neurochemical research and future in mainstream medicine. Ann N Y Acad Sci 2000; 909:186-216. [PMID: 10911931 DOI: 10.1111/j.1749-6632.2000.tb06683.x] [Citation(s) in RCA: 176] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In 1963, Professor Vincent P. Dole at the Rockefeller University formed a small team to develop a pharmacotherapy for the management of heroin addiction. They hypothesized that heroin addiction is a disease of the brain with behavioral manifestations, and not merely a personality disorder or criminal behavior and began to address the specific question of whether a long-acting opioid agonist could be used in the long-term maintenance treatment of heroin addiction. Over the next 35 years, many studies documented the safety, efficacy and effectiveness of methadone pharmacotherapy for heroin addiction, but Federal regulations and stigmatization of heroin addiction prevented implementation of treatment. Finally, in 1999, NIH published a report unequivocally supporting methadone maintenance pharmacotherapy for heroin addiction. Two other effective opioid agonist treatments have been developed: the even longer acting opioid agonist l-alpha-acetylmethadol (LAAM) has been approved for pharmacotherapy for heroin addiction, and still under study is the opioid partial agonist-antagonist buprenorphine-naloxone combination. A variety of studies, both laboratory based and clinical, have revealed the mechanisms of action of long-acting opioid agonists in treatment, including prevention of disruption of molecular, cellular and physiologic events and, in fact, allowing normalization of those functions disrupted by chronic heroin use. Recent molecular biological studies have revealed single nucleotide polymorphisms of the human mu opioid receptor gene; the mu opioid receptor is the site of action of heroin, the major opiate drug of abuse, analgesic agents such as morphine, and the major treatment agents for heroin addiction. These findings support the early hypotheses of our laboratory that addiction may be due to a combination of genetic, drug-induced and environmental (including behavioral) factors and also, that atypical stress responsivity may contribute to the acquisition and persistence of, as well as relapse to, use of addictive drugs.
Collapse
Affiliation(s)
- M J Kreek
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, New York 10021, USA.
| |
Collapse
|
794
|
Zuniga JR. Current Pain Research. Oral Maxillofac Surg Clin North Am 2000. [DOI: 10.1016/s1042-3699(20)30392-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
795
|
Li T, Liu X, Zhu ZH, Zhao J, Hu X, Sham PC, Collier DA. Association analysis of polymorphisms in the μ opioid gene and heroin abuse in Chinese subjects. Addict Biol 2000; 5:181-6. [PMID: 20575833 DOI: 10.1080/13556210050003775] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
We examined four polymorphisms in the μ opioid receptor gene in 282 Chinese heroin addicts from Sichuan Province, Southwest China and compared the allele and genotype frequencies to those in 258 normal controls from the same geographic region. Two of these polymorphisms (Ala6Val and Ser147Cys) were not polymorphic in the Chinese, with only Ala6 and Ser147 observed. The frequencies of the two other polymorphisms were significantly different from those observed in Caucasians, African Americans and Native Americans. The Asn40Asp and IVS2 + 691G/C polymorphisms did not differ significantly for allele (p= 0.16; p = 0.21), genotype (p= 0.32; p = 0.09) or haplotype frequencies (p= 0.24) between the Chinese heroin-addicted cases and normal controls. Similarly, we did not detect any association when the population was stratified by gender, route of administration (nasal inhalation and/or injection) and age-at-onset (above or below 25 years). This indicates that the μ opioid receptor is not likely to be a major genetic risk factor for heroin abuse in this population.
Collapse
|
796
|
Abstract
Alcohol withdrawal is a clinically and etiologically heterogeneous syndrome caused by a complex interaction of environmental (e.g., amount of ethanol) and genetic factors. Multiple genes are considered to be involved in various components of the syndrome, each of them contributing only modestly to withdrawal vulnerability. Association studies using candidate genes of the dopamine, serotonin, gabaergic and opioidergic systems are reviewed and methodological limitations are discussed.
Collapse
Affiliation(s)
- L G Schmidt
- Department of Psychiatry, Free University of Berlin, Eschenallee 3, 14050 Berlin, Germany
| | | |
Collapse
|
797
|
Abstract
Addiction to various substances, including drugs and alcohol, probably arises from a combination of environmental and genetic factors. The genetic vulnerability to drug addiction is supported by several familial, adoption and twin studies. However, as in other mental disorders, the genetic vulnerability to drug addiction appears complex: these disorders do not follow the rules of Mendelian inheritance. Instead, they are probably influenced by multiple susceptibility genes, each of which contributes to the disorder. The more genes necessary for a disorder, the harder it is to detect any of them. This difficulty is magnified by the role of environmental factors. Association studies using the candidate gene approach can identify susceptibility genes for drug abuse supported by the pathophysiological hypothesis of the illness. This review will focus on the clinical and molecular genetic studies in drug abuse.
Collapse
Affiliation(s)
- E Duaux
- Service Hospitalo-Universitaire de Sant¿e Mentale et Th¿erapeutique, H¿opital Sainte-Anne, 75014 Paris, France
| | | | | | | |
Collapse
|
798
|
Sander T, Berlin W, Gscheidel N, Wendel B, Janz D, Hoehe MR. Genetic variation of the human mu-opioid receptor and susceptibility to idiopathic absence epilepsy. Epilepsy Res 2000; 39:57-61. [PMID: 10690754 DOI: 10.1016/s0920-1211(99)00109-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Pharmacological and autoradiological studies suggest that mu-opioid receptor (OPRM) mediated neurotransmission is involved in the generation of absence seizures. Mutation screening of the human OPRM gene identified a common amino acid substitution polymorphism (Asn40Asp) that differentially modulates the binding affinity of beta-endorphin and signal transduction of the receptor. The present association study tested the candidate gene hypothesis that the Asn40Asp substitution polymorphism in the N-terminal OPRM domain confers genetic susceptibility to idiopathic absence epilepsy (IAE). The genotypes of the Asn40Asp polymorphism were assessed by allele-specific polymerase chain reaction in 72 German IAE patients and in 340 ethnically matched control subjects. The frequency of the Asp40 allele was significantly increased in the IAE patients [f(Asp40) = 0.139] compared to the controls [f(Asp40) = 0.078; chi2 = 5.467, df = 1, P = 0.019; OR = 2.03; 95%-CI: 1.12-3.68]. This allelic association suggests that the functional Asp40 variant of OPRM modulates neuronal excitability underlying the epileptogenesis of IAE.
Collapse
Affiliation(s)
- T Sander
- Department of Neurology, University Hospital Charité, Humboldt University of Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
799
|
Thome J, Gewirtz JC, Weijers HG, Wiesbeck GA, Henn FA. Genome polymorphism and alcoholism. Pharmacogenomics 2000; 1:63-71. [PMID: 11258598 DOI: 10.1517/14622416.1.1.63] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Different gene variants have been identified as risk or protective factors in alcoholism. The genes coding for dopamine receptors, serotonin transporters, and dehydrogenases represent susceptibility loci for addictive behaviour. However, alcoholism represents a complex psychiatric symptomatology which is caused by multiple factors, both genetic and environmental. Furthermore, there are probably different subtypes of alcoholism each with a distinct pathophysiology, and thus a different genetic background. Genetic research can help to identify such subtypes, which may require different therapeutic approaches. However, gene polymorphisms are not only responsible for a predisposition to alcoholism, but also for personality traits which influence the likelihood of developing addictive behaviour. Moreover, genetic polymorphisms are probably involved in the way an individual responds to treatment. Also, the severity of secondary diseases resulting from chronic alcohol uptake may depend on the genetic makeup of an individual. New treatment strategies focusing on genes contributing towards drug and alcohol dependence (such as gene therapy) are already under examination in animal models. However, further research is required before these developments will considerably change today's clinical handling of alcoholism.
Collapse
Affiliation(s)
- J Thome
- Laboratory of Biochemistry, Central Institute of Mental Health, Mannheim, Germany.
| | | | | | | | | |
Collapse
|
800
|
Abstract
Pharmacogenomics is defined as the study of the association between genetics and drug response. This is a rapidly expanding field with the hope that, within a few years, prospective genotyping will lead to patients being prescribed drugs which are both safer and more effective ('the right drug for the right patient', or personalized medicine). There are many existing examples in the literature of strong associations between genetic variation and drug response, and some of these even form the basis of accepted clinical tests. The molecular basis for some of these associations is described, and includes examples of variation in genes responsible for absorption and metabolism of the drug, and in target and disease genes. However, there are many issues surrounding the legal, regulatory and ethical framework to these studies that remain unanswered, and a huge amount of education both for the public and healthcare professionals will be needed before the results of this new medicine can be widely accepted.
Collapse
Affiliation(s)
- R March
- Research & Development Genetics, AstraZeneca, Mereside, Macclesfield, Cheshire, UK.
| |
Collapse
|