651
|
Bart G, LaForge KS, Borg L, Lilly C, Ho A, Kreek MJ. Altered levels of basal cortisol in healthy subjects with a 118G allele in exon 1 of the Mu opioid receptor gene. Neuropsychopharmacology 2006; 31:2313-7. [PMID: 16794569 DOI: 10.1038/sj.npp.1301128] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The mu opioid receptor is centrally involved in the development of the addictive diseases. It also modulates the stress responsive hypothalamic-pituitary-adrenal axis. Receptors encoded by the variant 118G polymorphism in exon 1 of the mu opioid receptor gene have a threefold increase in beta-endorphin binding and beta-endorphin is three times more potent in receptor-mediated activation of G protein-coupled inwardly rectifying potassium channels. Humans with this variant have increased stress response following opioid antagonism. Here, we study basal levels of adrenocorticotropic hormone and cortisol in subjects with this variant. In all, 59 healthy adults were genotyped and had morning levels of adrenocorticotropic hormone and cortisol measured following intravenous administration of saline placebo. Subjects with a 118G allele had significantly greater levels of cortisol than subjects with the prototype gene. Groups did not differ in levels of adrenocorticotropic hormone. A planned comparison revealed significantly greater cortisol in females with at least one copy of the 118G allele compared to females with the prototype gene. There was no significant effect of gender alone, nor was there a significant interaction between gender and genotype, on ACTH or cortisol. Subjects with at least one copy of the 118G allele have increased basal levels of cortisol, which may influence the susceptibility to and treatment of the stress responsive dyscrasia.
Collapse
Affiliation(s)
- Gavin Bart
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
652
|
Janicki PK, Schuler G, Francis D, Bohr A, Gordin V, Jarzembowski T, Ruiz-Velasco V, Mets B. A Genetic Association Study of the Functional A118G Polymorphism of the Human ??-Opioid Receptor Gene in Patients with Acute and Chronic Pain. Anesth Analg 2006; 103:1011-7. [PMID: 17000822 DOI: 10.1213/01.ane.0000231634.20341.88] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In this prospective, observational study we explored whether A118G single nucleotide polymorphism in the human mu-opioid receptor (MOR) gene could explain the inter-individual differences in opioid analgesic requirements in patients with acute postoperative pain and chronic pain. The frequency of the wild-type A118 MOR (major) and variant G118 MOR (minor) alleles in the subjects with chronic, noncancer pain (n = 121) and opioid-naïve subjects with acute postoperative pain (n = 101), serving as the control group, were examined. The relationships among the A118G MOR genotype, opioid requirements, and the numerical pain score were analyzed in both groups. The frequency of the minor allele was significantly lower in the subjects with chronic pain when compared with the group with acute postoperative pain (0.079 versus 0.158; P = 0.009 by chi2 test). No statistically significant association was observed between the presence of A118G MOR polymorphism and the average postoperative pain score or the doses of morphine used in the immediate postoperative period. In the high-quartile, opioid utilization, chronic pain patients, the homozygotic carriers of the major allele required significantly higher opioid dose than did the carriers of the minor allele. The results indicate that although the presence of the minor allele does not appear to affect opioid analgesic use in acute postoperative pain, the minor allele is less common in chronic pain patients, especially in those requiring higher doses of opioid analgesics.
Collapse
Affiliation(s)
- Piotr K Janicki
- Department of Anesthesiology, Pennsylvania State College of Medicine, Milton S. Hershey Medical Center, Hershey, Penn 17033-0850, USA.
| | | | | | | | | | | | | | | |
Collapse
|
653
|
Bruehl S, Chung OY. Parental history of chronic pain may be associated with impairments in endogenous opioid analgesic systems. Pain 2006; 124:287-294. [PMID: 16725261 DOI: 10.1016/j.pain.2006.04.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2006] [Revised: 03/28/2006] [Accepted: 04/17/2006] [Indexed: 11/16/2022]
Abstract
A family history of chronic pain has previously been linked to increased incidence of spontaneous acute pain and risk for chronic pain. Mechanisms underlying these associations are unknown, although similar effects on both acute and chronic pain suggest that central endogenous analgesic system differences may be relevant. This study tested whether a positive parental chronic pain history (PH+) was associated with impaired endogenous opioid analgesic responses to acute pain. Seventy-three chronic low back pain patients (LBP) and 46 pain-free controls received opioid blockade (8mg naloxone i.v.) and placebo blockade (saline) in randomized, counterbalanced order in separate sessions. During each, subjects participated in a 1-min finger pressure pain task followed by an ischemic forearm pain task, providing pain intensity ratings during and immediately following each task. To assess opioid analgesic function, blockade effects were derived by subtracting placebo from blockade condition pain responses. Placebo condition analyses indicated that both PH+ subjects and LBP subjects reported greater acute pain sensitivity than respective comparison groups (p's<.05). Multivariate analyses indicated that, beyond any influence of current chronic pain status, PH+ subjects failed to exhibit any endogenous opioid analgesia to acute ischemic pain, whereas PH- subjects elicited effective opioid analgesia (p<.05). A significant multivariate PHxSubject Type interaction (p<.05) indicated that opioid analgesic impairments were most prominent in PH+ LBP subjects. Similar analyses for finger pressure pain blockade effects were nonsignificant (p>.10). The possible heritability of endogenous opioid analgesic dysfunction observed in individuals with a positive parental chronic pain history remains to be investigated.
Collapse
Affiliation(s)
- Stephen Bruehl
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | |
Collapse
|
654
|
McGeary JE, Monti PM, Rohsenow DJ, Tidey J, Swift R, Miranda R. Genetic moderators of naltrexone's effects on alcohol cue reactivity. Alcohol Clin Exp Res 2006; 30:1288-96. [PMID: 16899031 DOI: 10.1111/j.1530-0277.2006.00156.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Naltrexone (NTX) reduces drinking and craving in alcoholic individuals in treatment and also in heavy drinkers. Polymorphisms in the D4 dopamine receptor (DRD4) gene and mu-opiate receptor gene (OPRM1) may moderate NTX's effects on craving. This study examined these candidate genes as moderators of the effects of NTX on cue-elicited urge to drink in non-treatment-seeking heavy drinkers. METHOD Data from the subset of 93 participants who consented for genetic testing in a larger study of medication effects were used to examine pharmacogenetic hypotheses. The non-treatment-seeking male and female heavy drinkers (62% alcohol dependent) were genotyped for the variable number of tandem repeats polymorphism in the DRD4 gene [L=7 or more (n=34), S=less than 7 (n=56)] and Asn40Asp single-nucleotide polymorphism in the OPRM1 gene [29 aspartate (Asp) carriers and 59 asparagine (Asn) homozygotes]. Ten days after randomization to NTX (50 mg) or placebo, participants completed an alcohol cue reactivity assessment. RESULTS Any medication effects were all accounted for by interaction with genotype. Naltrexone increased urge for alcohol in Asp carriers across alcohol and neutral beverage cue trials and had no effect on homozygous Asn carriers. Asp40 carriers on either medication had greater decreases (from resting baseline) in mean arterial blood pressure across all beverage cue trials compared with Asn carriers. For DRD4, no differential medication effects by DRD4 polymorphism were found. Alcohol dependence diagnosis did not moderate the effects of gene and medication on cue-elicited measures. DISCUSSION The differential responses to NTX due to variation in the OPRM1 gene may help explain conflicting results in clinical trials and suggest directions for patient-treatment matching.
Collapse
Affiliation(s)
- John E McGeary
- Center for Alcohol and Addiction Studies, Brown University, Providence, Rhode Island 02906, USA.
| | | | | | | | | | | |
Collapse
|
655
|
Nishizawa D, Han W, Hasegawa J, Ishida T, Numata Y, Sato T, Kawai A, Ikeda K. Association of mu-opioid receptor gene polymorphism A118G with alcohol dependence in a Japanese population. Neuropsychobiology 2006; 53:137-41. [PMID: 16679777 DOI: 10.1159/000093099] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2005] [Accepted: 02/14/2006] [Indexed: 11/19/2022]
Abstract
Ethanol is considered to activate the brain reward system by increasing the release of an endogenous opioid receptor ligand, beta-endorphin. The polymorphism A118G in the mu-opioid receptor gene (OPRM1) causes the amino acid change Asn40Asp and has been reported to affect the affinity of the ligand for the receptor. The association of this polymorphism with the vulnerability to alcohol dependence has been studied in many populations, but not yet in Japanese people. In the present study, we compared the frequencies of the polymorphism OPRM1 A118G between patients with alcohol dependence and healthy control subjects living in a Japanese provincial prefecture. We also genotyped a polymorphism, G1510A, in the acetaldehyde dehydrogenase 2 gene (ALDH2), in which the A allele causes poor metabolism of acetaldehyde, a major metabolite of alcohol. Both OPRM1 118G and ALDH2 1510G were significantly associated with alcohol dependence. These results suggest that OPRM1 118G in addition to ALDH2 1510G might be one of the risk factors for alcohol dependence in Japanese people.
Collapse
Affiliation(s)
- Daisuke Nishizawa
- Division of Psychobiology, Tokyo Institute of Psychiatry, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
656
|
Oslin DW, Berrettini WH, O'Brien CP. Targeting treatments for alcohol dependence: the pharmacogenetics of naltrexone. Addict Biol 2006; 11:397-403. [PMID: 16961767 DOI: 10.1111/j.1369-1600.2006.00036.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Alcohol dependence is one of the leading causes of morbidity worldwide, yet only a minority of those afflicted engages in treatment. While increasing access to treatment is an important public health approach, increasing the success of treatment is also likely to lead to greater engagement. However, alcohol dependence is a complex disorder likely to consist of several biological subtypes. Recent evidence from a number of different studies has suggested that genetic variation in the mu-opioid receptor has a significant influence on clinical presentation of alcohol problems and response to treatment with an opioid antagonist. Most notably, the A118G polymorphism of the mu-receptor gene has been demonstrated to predict clinical response to naltrexone in alcohol-dependent individuals. This article reviews the biological correlates and outlines a scientific agenda for better understanding the role of opioid neurotransmission in the etiology, maintenance and treatment of alcohol dependence.
Collapse
Affiliation(s)
- David W Oslin
- University of Pennsylvania, Geriartric and Addiction Psychiatry, philadelphia, PA 19104, USA
| | | | | |
Collapse
|
657
|
Trujillo KA, Castañeda E, Martínez D, González G. Biological research on drug abuse and addiction in Hispanics: current status and future directions. Drug Alcohol Depend 2006; 84 Suppl 1:S17-28. [PMID: 16777354 DOI: 10.1016/j.drugalcdep.2006.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Impressive progress has been made in the understanding of biological contributions to drug abuse and addiction. An area that has only recently begun to receive attention is potential ethnic and racial differences in biological systems that contribute to, or protect from, problem drug use. This article reviews recent research on drug abuse and addiction in Hispanics in which biological questions have been addressed, including work on genes, gene products (proteins), physiology and pharmacotherapy. Taken together, work to date suggests that there are both similarities and differences between Hispanics and other ethnic groups in biological factors related to drug abuse and addiction. Although the results are intriguing, relatively few studies have been done, and those that have been done have often been inconclusive due to low numbers of Hispanic subjects. Moreover, studies have often failed to recognize the complexity and heterogeneity of Hispanic populations in the United States and around the world. After reviewing the current status of the field, recommendations are given for future research in both humans and relevant animal models that will lead to a better understanding of drug abuse and addiction in Hispanics.
Collapse
Affiliation(s)
- Keith A Trujillo
- Department of Psychology and Office for Biomedical Research and Training, California State University San Marcos, San Marcos, CA 92096, USA.
| | | | | | | |
Collapse
|
658
|
|
659
|
Wang D, Sadée W. Searching for polymorphisms that affect gene expression and mRNA processing: example ABCB1 (MDR1). AAPS JOURNAL 2006; 8:E515-20. [PMID: 17025270 PMCID: PMC2761059 DOI: 10.1208/aapsj080361] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cis-acting genetic variations can affect the amount and structure of mRNA/protein. Genomic surveys indicate that polymorphisms affecting transcription and mRNA processing, including splicing and turnover, may account for the main share of genetic factors in human phenotypic variability; however, most of these polymorphisms remain yet to be discovered. We use allelic expression imbalance (AEI) as a quantitative phenotype in the search for functional cis-acting polymorphisms in many genes, including ABCB1 (multidrug resistance 1 gene, MDR1, Pgp). Previous studies have shown that ABCB1 activity correlates with a synonymous polymorphism, C3435T; however, the functional polymorphism and molecular mechanism underlying this clinical association remained unknown. Analysis of allele-specific expression in liver autopsy samples and in vitro expression experiments showed that C3435T represents a main functional polymorphism, accounting for 1.5- to 2-fold changes in mRNA levels. The mechanism appears to involve increased mRNA turnover, probably as a result of different folding structures calculated for mRNA with the Mfold program. Other examples of the successful application of AEI analysis for studying functional polymorphism include 5-HTT (serotonin transporter, SLC6A4) and OPRM1 (mu opioid receptor). AEI is therefore a powerful approach for detecting cis-acting polymorphisms affecting gene expression and mRNA processing.
Collapse
Affiliation(s)
- Danxin Wang
- Department of Pharmacology, Program in Pharmacogenetics, College of Medicine and Public Health, The Ohio State University, 333 West 10th Ave, Graves Hall 5168, Columbus, OH 43210, USA.
| | | |
Collapse
|
660
|
Zhang L, Kendler KS, Chen X. The mu-opioid receptor gene and smoking initiation and nicotine dependence. Behav Brain Funct 2006; 2:28. [PMID: 16887046 PMCID: PMC1557520 DOI: 10.1186/1744-9081-2-28] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2006] [Accepted: 08/04/2006] [Indexed: 11/10/2022] Open
Abstract
The gene encoding the mu-opioid receptor (OPRM1) is reported to be associated with a range of substance dependence. Experiments in knockout mice indicate that the mu-opioid receptor may mediate reinforcing effects of nicotine. In humans, opioid antagonist naltrexone may reduce the reinforcing effects of tobacco smoking. Additionally, the OPRM1 gene is located in a region showing linkage to nicotine dependence. The OPRM1 is thus a plausible candidate gene for smoking behavior. To investigate whether OPRM1 contributes to the susceptibility of smoking initiation and nicotine dependence, we genotyped 11 SNPs in the gene for 688 Caucasian subjects of lifetime smokers and nonsmokers. Three SNPs showed nominal significance for smoking initiation and one reached significance for nicotine dependence. The global test for three-marker (rs9479757-rs2075572-rs10485057) haplotypes was significant for smoking initiation (p = 0.0022). The same three-marker haplotype test was marginal (p = 0.0514) for nicotine dependence. These results suggest that OPRM1 may be involved in smoking initiation and nicotine dependence.
Collapse
Affiliation(s)
- Lan Zhang
- Virginia Institute of Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA 23298, USA
- Psychiatry Department, West China Hospital, Sichuan University, Chengdu, China
| | - Kenneth S Kendler
- Virginia Institute of Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Xiangning Chen
- Virginia Institute of Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
661
|
Chou WY, Yang LC, Lu HF, Ko JY, Wang CH, Lin SH, Lee TH, Concejero A, Hsu CJ. Association of mu-opioid receptor gene polymorphism (A118G) with variations in morphine consumption for analgesia after total knee arthroplasty. Acta Anaesthesiol Scand 2006; 50:787-92. [PMID: 16879459 DOI: 10.1111/j.1399-6576.2006.01058.x] [Citation(s) in RCA: 203] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Morphine consumption after a given surgical procedure can vary considerably. Studies show that single nucleotide polymorphism involving the nucleotide position 118 at exon 1 of the mu-opioid receptor gene (OPRM1) may play a role in mediating the effects of opioids. This study was performed to correlate the A118G polymorphism at OPRM1 with morphine consumption in patients undergoing total knee arthroplasty. METHODS Post-operative pain was relieved by patient-controlled analgesia (PCA). The analgesic effect was evaluated using a visual analogue scale. Side-effects, such as sedation, nausea and vomiting, and pruritus, were recorded systematically. The genotypes were determined by sequencing polymerase chain reaction-amplified DNA. The differences in demographics and consumed morphine from the PCA device between the different genotypes were tested using one-way analysis of variance. The prevalence of side-effects from morphine and sex distribution were compared using the Kruskal-Wallis test. RESULTS One hundred and forty-seven patients were included in the study. Twenty-seven patients who required rescue analgesia were excluded; these patients did not differ demographically or genetically from the 120 who completed the study. Of the latter, 74 were A118 homozygous (AA), 33 were heterozygous (AG) and 13 were G118 homozygous (GG). Group GG consumed significantly more morphine (40.4 +/- 22.0 mg) than group AA (25.3 +/- 15.5 mg) and group AG (25.6 +/- 11.7 mg) during the first 48 h post-operatively. The groups did not differ with respect to reported pain, age, sex, weight and adverse effects. CONCLUSIONS G118 homozygotes have a poorer response to morphine for post-operative pain control than A118 homozygotes or heterozygotes. The genotype may thus influence the post-operative response to pain and cause differences in the amounts of analgesic consumed by patients after total knee arthroplasty.
Collapse
Affiliation(s)
- W-Y Chou
- Department of Anaesthesiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
662
|
Lötsch J, Geisslinger G. Relevance of frequent mu-opioid receptor polymorphisms for opioid activity in healthy volunteers. THE PHARMACOGENOMICS JOURNAL 2006; 6:200-10. [PMID: 16415919 DOI: 10.1038/sj.tpj.6500362] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Polymorphisms in the mu-opioid receptor gene (OPRM1) are primary candidate sources of clinical variability in opioid therapy. Apart from the 118A>G single nucleotide polymorphism, nothing is known about the role of OPRM1 mutations in opioid therapy. The influence of the OPRM1 mutations on opioid pharmacodynamics was assessed in pooled data from 31 healthy volunteers obtained in previous studies with available plasma concentrations and pupil diameters after intravenous administration of morphine or morphine-6-glucuronide (M6G). A total of 24 candidate ORPM1 mutations were screened for and those found at an allelic frequency of at least 5% in the 31 subjects were analyzed for functional consequences, using population pharmacokinetic-pharmacodynamic modeling of the miotic effects of the opioids as a reliable and sensitive surrogate parameter of the central nervous opioid effects. Polymorphisms at an allelic frequency of > or =5% (n=310) were 118A>G in exon 1 (11.5%), the IVS2-31G>A (8.9%) and IVS2-691C>G (44.5%) SNPs in intron 2. The 118A>G SNP significantly increased the values of EC50 by a factor of more than 2 (non-mutated: EC50,morphine=30 nmol/l, EC50,M6G=750 nmol/l, 118G carriers: EC50,morphine=66 nmol/l, EC50,M6G=1650 nmol/l), whereas the IVS2-691C>G SNP had no effect. Based on morphine and M6G, the present analysis encourages focusing on the 118A>G SNP when investigating the role of OPRM1 mutations for the activity of opioid analgesics. Other OPRM1 mutations are probably less important either owing to low allelic frequency or due to poor indications for functional consequences. This applies to opioid potency in the context of opioid therapy but not to pain processing or substance addiction, in which opioid receptors are involved but other or additional OPRM1 mutations may be important.
Collapse
Affiliation(s)
- J Lötsch
- pharmazentrum frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany.
| | | |
Collapse
|
663
|
Ide S, Kobayashi H, Ujike H, Ozaki N, Sekine Y, Inada T, Harano M, Komiyama T, Yamada M, Iyo M, Iwata N, Tanaka K, Shen H, Iwahashi K, Itokawa M, Minami M, Satoh M, Ikeda K, Sora I. Linkage disequilibrium and association with methamphetamine dependence/psychosis of mu-opioid receptor gene polymorphisms. THE PHARMACOGENOMICS JOURNAL 2006; 6:179-88. [PMID: 16402083 DOI: 10.1038/sj.tpj.6500355] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Several studies indicate that the mu-opioid receptor plays a role in addiction not only to opiate drugs but also to alcohol and non-opiate addictive drugs. Our studies aim to reveal the associations between gene polymorphisms and methamphetamine (MAP) dependence/psychosis. We newly identified several polymorphisms and four substantial linkage disequilibrium (LD) blocks in the mu-opioid receptor (OPRM1) gene. We found significant differences in both genotype and allele frequencies of the single-nucleotide polymorphism (SNP) IVS2+G691C between control (n=232) and MAP-dependent/psychotic patients (n=128). There was also a significant association between IVS2+G691C and patients with transient psychosis. These results suggest that the OPRM1 gene variations may be a factor in development and prognosis of MAP psychosis.
Collapse
Affiliation(s)
- S Ide
- Division of Psychobiology, Tokyo Institute of Psychiatry, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
664
|
Kampov-Polevoy AB, Alterman A, Khalitov E, Garbutt JC. Sweet preference predicts mood altering effect of and impaired control over eating sweet foods. Eat Behav 2006; 7:181-7. [PMID: 16843219 DOI: 10.1016/j.eatbeh.2005.09.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2005] [Revised: 09/06/2005] [Accepted: 09/20/2005] [Indexed: 10/25/2022]
Abstract
The purpose of the present study was to examine association between hedonic response to sweet taste and a mood altering effect associated with eating sweet foods and impaired control over eating sweets. Participants (n=163, 39% males) rated a series of sucrose solutions for intensity of sweetness and palatability and completed a newly developed 12-item Sweet Taste Questionnaire (STQ). It was shown that STQ identifies two factors in the individual's attitude towards sweet foods: sensitivity to the mood altering effect of sweets and impaired control over eating sweet foods. Individuals preferring the taste of the strongest offered sucrose concentration reported a stronger mood altering effect associated with eating of sweet foods and were more likely to have an impaired control over eating sweets than the rest of the group. Women generally had higher scores on both factors compared to men. The results of the present study support the hypothesis that hedonic response to sweet taste is associated with elevated sensitivity to mood altering effects of sweet foods and impaired control over eating sweets.
Collapse
Affiliation(s)
- Alexey B Kampov-Polevoy
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7160, USA.
| | | | | | | |
Collapse
|
665
|
Samer CF, Desmeules JA, Dayer P. Individualizing analgesic prescription Part I: pharmacogenetics of opioid analgesics. Per Med 2006; 3:239-269. [PMID: 29788657 DOI: 10.2217/17410541.3.3.239] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The current use of analgesics is based on the empiric administration of a given drug with clinical monitoring for efficacy and toxicity. However, individual responses to drugs are influenced by a combination of pharmacokinetic and pharmacodynamic processes, and each of these components, in addition to pain perception and processing, seem to be regulated by genetic factors. Whereas polymorphic drug-metabolizing enzymes and drug transporters may affect the pharmacokinetics of drugs, polymorphic drug targets and disease-related pathways may influence the pharmacodynamic action of drugs. After usual dose, drug toxicity, as well as inefficacy, can be observed depending on the polymorphism, the analgesic considered and the presence or absence of active metabolites. Thus, cytochrome P450 (CYP)2D6 polymorphism influences codeine and tramadol analgesic effects, CYP2C9 has an impact on the disposition of some nonsteroidal anti-inflammatory drugs, and opioid receptor polymorphism (118A>G) may reduce morphine potency. Moreover, drug interaction mimics genetic deficiency and contributes to the variability in response to analgesics. This two-part review summarizes the available data on the pharmacokinetic-pharmacodynamic consequences of known polymorphisms of drug-metabolizing enzymes (CYP and uridine diphosphate glucuronosyltransferase), drug transporters (multidrug resistance proteins, multidrug resistance-associated proteins, organic anion-transporting polypeptides, and serotonin transporters), relevant drug targets (such as µ-opioid receptor, serotonin receptor and cyclooxygenases) and other nonopioid biological systems, on currently prescribed central and peripheral analgesics.
Collapse
Affiliation(s)
- Caroline Flora Samer
- Geneva University Hospitals, Clinical Pharmacology and Toxicology and Multidisciplinary Pain Centre, Department of Anaesthesiology, Pharmacology and Intensive Care, 1211 Geneva 14, Switzerland.
| | - Jules Alexandre Desmeules
- Geneva University Hospitals, Clinical Pharmacology and Toxicology and Multidisciplinary Pain Centre, Department of Anaesthesiology, Pharmacology and Intensive Care, 1211 Geneva 14, Switzerland.
| | - Pierre Dayer
- Geneva University Hospitals, Clinical Pharmacology and Toxicology and Multidisciplinary Pain Centre, Department of Anaesthesiology, Pharmacology and Intensive Care, 1211 Geneva 14, Switzerland.
| |
Collapse
|
666
|
Kumar R, Orsoni S, Norman L, Verma AS, Tirado G, Giavedoni LD, Staprans S, Miller GM, Buch SJ, Kumar A. Chronic morphine exposure causes pronounced virus replication in cerebral compartment and accelerated onset of AIDS in SIV/SHIV-infected Indian rhesus macaques. Virology 2006; 354:192-206. [PMID: 16876224 DOI: 10.1016/j.virol.2006.06.020] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2006] [Revised: 06/14/2006] [Accepted: 06/19/2006] [Indexed: 10/24/2022]
Abstract
Six morphine-exposed and 3 control male Indian rhesus macaques were intravenously inoculated with mixture of SHIV(KU), SHIV(89.6)P and SIV/17E-Fr. These animals were followed for a period of 56 weeks in order to determine CD4 and CD8 profile, viral loads in plasma and cerebrospinal fluid (CSF), relative distribution of 3 pathogenic viruses in blood and brain, binding as well neutralizing antibody levels and cellular immune responses. Both morphine-exposed and control macaques showed a precipitous loss of CD4+ T cells; control animals, however, showed a greater tendency to recover these cells than did their morphine-exposed counterparts. The plasma and CSF viral loads were significantly higher in morphine-exposed group than those in the control group. Four morphine-exposed animals succumbed to SIV/SHIV-induced AIDS at week 18, 19, 20 and 51; post-infection with neurological disorders was found in 3 of the 4 animals. At the end of the 56-week observation period, 2 morphine-exposed and 3 control animals were still alive. All 3 viruses replicated in the blood of both morphine-exposed and control macaques, but the cerebral compartment showed a selection phenomenon; only SIV/17E-Fr and SHIV(KU) successfully crossed the blood brain barrier (BBB). The morphine-exposed macaques further favored viral migration through the blood brain barrier (BBB). SIV/17E-Fr crossed the BBB within 2 weeks in both morphine-exposed and control macaques, whereas SHIV(KU) crossed the BBB more rapidly in morphine-exposed than in control macaques. Three morphine-exposed macaques (euthanized at weeks 18, 19 and 20) did not develop cellular or humoral immune responses, whereas the other 3 morphine-exposed and 3 control macaques developed both cellular and humoral immune responses.
Collapse
Affiliation(s)
- Rakesh Kumar
- Laboratory of Viral Immunology, AIDS Research Program and Department of Microbiology, Ponce School of Medicine, Ponce, PR 00732, Puerto Rico
| | | | | | | | | | | | | | | | | | | |
Collapse
|
667
|
Arias A, Feinn R, Kranzler HR. Association of an Asn40Asp (A118G) polymorphism in the mu-opioid receptor gene with substance dependence: a meta-analysis. Drug Alcohol Depend 2006; 83:262-8. [PMID: 16387451 DOI: 10.1016/j.drugalcdep.2005.11.024] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2005] [Revised: 11/02/2005] [Accepted: 11/28/2005] [Indexed: 11/20/2022]
Abstract
INTRODUCTION The mu-opioid receptor has been implicated in the pathogenesis of dependence on opioids, alcohol, nicotine, and cocaine. Studies examining the association of the mu-opioid receptor gene (genetic locus OPRM1) with substance dependence (SD) have focused on the Asn40Asp (A118G) single nucleotide polymorphism (SNP). METHOD We used meta-analysis to examine the literature on the association of Asn40Asp with SD. Twenty-two articles describing 28 distinct samples and over 8000 subjects were included. A variety of factors (i.e., ethnicity, type of SD, rigor with which controls were screened, severity of SD among cases) were examined as potential moderators of the association. RESULTS Four studies showed a significantly higher frequency of the Asp40 allele among SD cases, while three studies showed a significantly higher frequency of the Asp40 allele among controls. There was no significant association between Asn40Asp and SD (OR=1.01, 95%CI=0.86-1.19), nor was there substantial evidence of a moderator effect. CONCLUSION The Asn40Asp SNP in OPRM1 does not appear to affect risk for SD. Additional research is needed to determine whether these findings reflect no role for OPRM1 in determining risk for SD or whether another polymorphism in the gene influences receptor function and risk for SD.
Collapse
Affiliation(s)
- Albert Arias
- Alcohol Research Center, Department of Psychiatry, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030-2103, USA
| | | | | |
Collapse
|
668
|
Bayerer B, Stamer U, Hoeft A, Stüber F. Genomic variations and transcriptional regulation of the human mu-opioid receptor gene. Eur J Pain 2006; 11:421-7. [PMID: 16843022 DOI: 10.1016/j.ejpain.2006.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Revised: 03/26/2006] [Accepted: 06/03/2006] [Indexed: 11/25/2022]
Abstract
The mu-opioid receptor (MOR1) is a target of endogenous and exogenous opioids and plays a pivotal role for anesthesia and analgesia. Variations in the 5' flanking sequence of the mu-opioid receptor gene may influence transcriptional regulation and ultimately alter protein expression of MOR1. In the present study we investigated the influence of eight single nucleotide polymorphisms (SNP) within the mu-opioid receptor promoter on promoter activity and evaluated the frequencies of the relevant SNPs in 700 patients under opioid medication. Reporter-gene-constructs were created by means of PCR and site directed mutagenesis, testing eight SNPs previously described. The neuroblastoma cell line SHSY5Y was used for transfection and promoter activity was estimated by luciferase activity. Of the eight reporter gene constructs employed to test genomic variations, two produced a significant change in luciferase activity when compared to wild-type constructs. The G-554A variation located within a known NFkB binding element resulted in a decreased activity whereas the A/G base exchange at position -1320 showed an increased luciferase activity. This particular variant generated a myeloid zinc finger (MZF1) cis-acting element known to impact transcription. The allele frequency of the -1320G variant was 0.21% in 700 Caucasian patients under opioid medication in contrast to 9.1% reported previously in drug addicted African Americans. Because of this unexpected low frequency an association analysis to opioid requirements and effects of mu-opioid receptor agonists was not feasible. In conclusion, transcriptional regulation of MOR1 is modified by two genetic variations at positions -554 and -1320 of the mu-opioid receptor promoter. Individuals presenting these variations may have an altered level of MOR expression. A possible association of these genomic variants on efficacy and side effects of opioid treatment in different ethnic groups has to be elucidated.
Collapse
Affiliation(s)
- Bettina Bayerer
- Department of Anesthesiology and Intensive Care Medicine, University of Bonn, Sigmund-Freud-Strasse 25, 53105 Bonn, Germany
| | | | | | | |
Collapse
|
669
|
Kirchheiner J, Schmidt H, Tzvetkov M, Keulen JTHA, Lötsch J, Roots I, Brockmöller J. Pharmacokinetics of codeine and its metabolite morphine in ultra-rapid metabolizers due to CYP2D6 duplication. THE PHARMACOGENOMICS JOURNAL 2006; 7:257-65. [PMID: 16819548 DOI: 10.1038/sj.tpj.6500406] [Citation(s) in RCA: 256] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Codeine is an analgesic drug acting on mu-opiate receptors predominantly via its metabolite morphine, which is formed almost exclusively by the genetically polymorphic enzyme cytochrome P450 2D6 (CYP2D6). Whereas it is known that individuals lacking CYP2D6 activity (poor metabolizers, PM) suffer from poor analgesia from codeine, ultra-fast metabolizers (UM) due to the CYP2D6 gene duplication may experience exaggerated and even potentially dangerous opioidergic effects and no systematical study has been performed so far on this question. A single dose of 30 mg codeine was administered to 12 UM of CYP2D6 substrates carrying a CYP2D6 gene duplication, 11 extensive metabolizers (EM) and three PM. Genotyping was performed using polymerase chain reaction-restriction fragment length polymorphism methods and a single-base primer extension method for characterization of the gene-duplication alleles. Pharmacokinetics was measured over 24 h after drug intake and codeine and its metabolites in plasma and urine were analyzed by liquid chromatography with tandem mass spectrometry. Significant differences between the EM and UM groups were detected in areas under the plasma concentration versus time curves (AUCs) of morphine with a median (range) AUC of 11 (5-17) microg h l(-1) in EMs and 16 (10-24) microg h l(-1) in UM (P=0.02). In urine collected over 12 h, the metabolic ratios of the codeine+codeine-6-glucuronide divided by the sum of morphine+its glucuronides metabolites were 11 (6-17) in EMs and 9 (6-16) in UM (P=0.05). Ten of the 11 CYP2D6 UMs felt sedation (91%) compared to six (50%) of the 12 EMs (P=0.03). CYP2D6 genotypes predicting ultrarapid metabolism resulted in about 50% higher plasma concentrations of morphine and its glucuronides compared with the EM. No severe adverse effects were seen in the UMs in our study most likely because we used for safety reasons a low dose of only 30 mg. It might be good if physicians would know about the CYP2D6 duplication genotype of their patients before administering codeine.
Collapse
Affiliation(s)
- J Kirchheiner
- Department of Pharmacology of Natural Products and Clinical Pharmacology, University Ulm, Ulm, Germany.
| | | | | | | | | | | | | |
Collapse
|
670
|
Abstract
Genomic variations influencing nociceptive sensitivity and susceptibility to pain conditions, as well as responses to pharmacotherapy of pain are currently under investigation. Candidate genes involved in pain perception, pain processing and pain management such as (opioid) receptors, transporters and other targets of pharmacotherapy are discussed. Drug metabolizing enzymes represent a further major target of ongoing research in order to identify associations between an individual's genetic profile and drug response (pharmacogenetics). Polymorphisms of the cytochrome P 450 enzymes influence analgesic efficacy of codeine, tramadol and tricyclic antidepressants (CYP2D6). Blood levels of some non-steroidal anti-inflammatory drugs (NSAIDs) are dependent on CYP2C9 activity, whereas opioid receptor polymorphisms are discussed with respect to differences in opioid-mediated analgesia and side-effects. Pharmacogenetics is seen as a potential diagnostic tool for improving patient therapy and care and will contribute to a more individualized drug treatment in the future.
Collapse
Affiliation(s)
- U Stamer
- Klinik und Poliklinik für Anästhesiologie und Operative Intensivmedizin, Rheinische Friedrich-Wilhelms-Universität, Sigmund-Freud-Str. 25, 53105, Bonn.
| | | | | |
Collapse
|
671
|
Ross JR, Riley J, Quigley C, Welsh KI. Clinical Pharmacology and Pharmacotherapy of Opioid Switching in Cancer Patients. Oncologist 2006; 11:765-73. [PMID: 16880235 DOI: 10.1634/theoncologist.11-7-765] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Pain is one of the most common and often most feared symptoms in patients with cancer. Ongoing or progressive pain is physically debilitating and has a marked impact on quality of life. Since a third of the population will die from cancer, and of these, 80% will experience severe pain in their final year of life, effective treatment of cancer-related pain remains both a high priority and an ongoing challenge in clinical practice. Individuals with moderate to severe cancer-related pain require treatment with strong analgesics, namely opioids. There is evidence to support the therapeutic maneuver of opioid switching in clinical practice, but further evidence is needed to elucidate the underlying mechanisms for interindividual differences in response to different opioids. Large, robust clinical trials will be needed if clinical differences among side-effect profiles of different opioids are to be clearly demonstrated. This review discusses candidate genes, which contribute to opioid response; many other genes have also been implicated in "pain" from animal or human studies. In order to continue to evaluate the genetic contributions to both pain susceptibility and analgesic response, further candidate genes need to be considered. Good pain control remains a high priority for clinicians and patients, and there is much work to be done to further individualize analgesic therapy for patients with cancer.
Collapse
Affiliation(s)
- Joy R Ross
- Department of Palliative Medicine, Horder Ward, Royal Marsden Hospital, London SW3 6JJ, United Kingdom.
| | | | | | | |
Collapse
|
672
|
Lee AM, Tyndale RF. Drugs and genotypes: how pharmacogenetic information could improve smoking cessation treatment. J Psychopharmacol 2006; 20:7-14. [PMID: 16785264 DOI: 10.1177/1359786806066039] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Current smoking cessation treatments are not optimal as the long-term abstinence rates are low. Nicotine replacement therapy and bupropion are the only pharmacotherapies widely available to smokers and there is a need to improve current cessation treatments and to develop new drug therapies. Our goal is to use pharmacogenetic information to improve smoking cessation treatments. Candidate genes include those involved in central mechanisms (such as genes encoding the nicotinic acetylcholine receptors, dopamine receptors, dopamine transporters and opioid receptors) and peripheral mechanisms (such as genes encoding the drug-metabolizing enzymes CYP2A6 and CYP2B6). Pharmacogenetics could be used to improve current smoking cessation treatments through two general approaches. The first would be to identify novel drug targets based on genetic variants that are associated with a natural protection against nicotine dependence, decreased smoking behaviour and/or increased smoking cessation. This knowledge could be used to develop drugs that can mimic these advantages, reducing the risk for acquisition, block the rewarding effects of smoking, decreasing the amount smoked and increasing cessation. The second would be to identify smokers with particular genetic variants who are likely to respond better to one treatment (or dose) versus another and to individualize their treatment to maximize long-term abstinence. This review will highlight examples of how pharmacogenetic information from central and peripheral candidate genes could contribute to improving smoking cessation outcomes by these two approaches.
Collapse
Affiliation(s)
- Anna M Lee
- Centre for Addiction and Mental Health, Department of Pharmacology, University of Toronto, Toronto, Canada
| | | |
Collapse
|
673
|
Bruehl S, Chung OY, Burns JW. Anger expression and pain: an overview of findings and possible mechanisms. J Behav Med 2006; 29:593-606. [PMID: 16807797 DOI: 10.1007/s10865-006-9060-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2006] [Indexed: 11/28/2022]
Abstract
A tendency to manage anger via direct expression (anger-out) is increasingly recognized as influencing responses to pain. Elevated trait anger-out is associated with increased responsiveness to acute experimental and clinical pain stimuli, and is generally related to elevated chronic pain intensity in individuals with diverse pain conditions. Possible mechanisms for these links are explored, including negative affect, psychodynamics, central adipose tissue, symptom specific muscle reactivity, endogenous opioid dysfunction, and genetics. The opioid dysfunction hypothesis has some experimental support, and simultaneously can account for anger-out's effects on both acute and chronic pain. Factors which may moderate the anger-out/pain link are described, including narcotic use, gender, and genetic polymorphisms. Pain exacerbating effects of trait anger-out are contrasted with the apparent pain inhibitory effects of behavioral anger expression exhibited in anger-provoking contexts. Conceptual issues related to the state versus trait effects of expressive anger regulation are discussed.
Collapse
Affiliation(s)
- Stephen Bruehl
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37212, USA.
| | | | | |
Collapse
|
674
|
Munafò MR, Shields AE, Berrettini WH, Patterson F, Lerman C. Pharmacogenetics and nicotine addiction treatment. Pharmacogenomics 2006; 6:211-23. [PMID: 16013953 DOI: 10.1517/14622416.6.3.211] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
This review focuses on the current status of, and future directions for, pharmacogenetic research on nicotine dependence and smoking cessation treatment. Pharmacological treatment involving nicotine replacement therapy and bupropion for nicotine addiction and smoking cessation has been shown to be efficacious when provided in combination with behavioral support. Cessation rates remain somewhat modest, however, and one possibility is that success rates may be enhanced by offering treatments tailored to an individual's genotype. Nonetheless, research on this issue remains in its infancy, and although the scope for individualized treatment tailored to genotype is promising, there are substantial practical, ethical and social considerations that must be addressed before such research is translated into clinical practice.
Collapse
Affiliation(s)
- Marcus R Munafò
- University of Bristol, Department of Experimental Psychology, Bristol BS8 ITN, UK.
| | | | | | | | | |
Collapse
|
675
|
Smith YR, Stohler CS, Nichols TE, Bueller JA, Koeppe RA, Zubieta JK. Pronociceptive and antinociceptive effects of estradiol through endogenous opioid neurotransmission in women. J Neurosci 2006; 26:5777-85. [PMID: 16723535 PMCID: PMC1808228 DOI: 10.1523/jneurosci.5223-05.2006] [Citation(s) in RCA: 246] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Prominent interindividual and sex-dependent differences have been described in responses to sustained pain and other stressful stimuli. Variations in mu-opioid receptor-mediated endogenous opioid neurotransmission may underlie some of these processes. We examined both baseline mu-opioid receptor levels and the activation of this neurotransmitter system during sustained pain using positron emission tomography in a sample of young healthy men and women. Women were studied twice, during low and high estrogen states. The high-estrogen state was associated with regional increases in baseline mu-opioid receptor availability in vivo and a greater activation of endogenous opioid neurotransmission during the pain stressor. The latter did not differ from that obtained in males. During the low estrogen condition, however, significant reductions in endogenous opioid tone were observed at the level of thalamus, nucleus accumbens, and amygdala, which were associated with hyperalgesic responses. Estrogen-associated variations in the activity of mu-opioid neurotransmission correlated with individual ratings of the sensory and affective perceptions of the pain and the subsequent recall of that experience. These data demonstrate a significant role of estrogen in modulating endogenous opioid neurotransmission and associated psychophysical responses to a pain stressor in humans.
Collapse
|
676
|
van Dorp ELA, Romberg R, Sarton E, Bovill JG, Dahan A. Morphine-6-Glucuronide: Morphine??s Successor for Postoperative Pain Relief? Anesth Analg 2006; 102:1789-97. [PMID: 16717327 DOI: 10.1213/01.ane.0000217197.96784.c3] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In searching for an analgesic with fewer side effects than morphine, examination of morphine's active metabolite, morphine-6-glucuronide (M6G), suggests that M6G is possibly such a drug. In contrast to morphine, M6G is not metabolized but excreted via the kidneys and exhibits enterohepatic cycling, as it is a substrate for multidrug resistance transporter proteins in the liver and intestines. M6G exhibits a delay in its analgesic effect (blood-effect site equilibration half-life 4-8 h), which is partly related to slow passage through the blood-brain barrier and distribution within the brain compartment. In humans, M6G's potency is just half of that of morphine. In clinical studies, M6G is well tolerated and produces adequate and long lasting postoperative analgesia. At analgesic doses, M6G causes similar reduction of the ventilatory response to CO2 as an equianalgesic dose of morphine but significantly less depression of the hypoxic ventilatory response. Preliminary data indicate that M6G is associated less than morphine with nausea and vomiting, causing 50% and 75% less nausea in postoperative and experimental settings, respectively. Although the data from the literature are very promising, we believe that more studies are necessary before we may conclude that M6G is superior to morphine for postoperative analgesia.
Collapse
Affiliation(s)
- Eveline L A van Dorp
- Department of Anesthesiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
677
|
Drakenberg K, Nikoshkov A, Horváth MC, Fagergren P, Gharibyan A, Saarelainen K, Rahman S, Nylander I, Bakalkin G, Rajs J, Keller E, Hurd YL. Mu opioid receptor A118G polymorphism in association with striatal opioid neuropeptide gene expression in heroin abusers. Proc Natl Acad Sci U S A 2006; 103:7883-8. [PMID: 16682632 PMCID: PMC1472539 DOI: 10.1073/pnas.0600871103] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Indexed: 01/25/2023] Open
Abstract
Mu opioid receptors are critical for heroin dependence, and A118G SNP of the mu opioid receptor gene (OPRM1) has been linked with heroin abuse. In our population of European Caucasians (n = 118), approximately 90% of 118G allelic carriers were heroin users. Postmortem brain analyses showed the OPRM1 genotype associated with transcription, translation, and processing of the human striatal opioid neuropeptide system. Whereas down-regulation of preproenkephalin and preprodynorphin genes was evident in all heroin users, the effects were exaggerated in 118G subjects and were most prominent for preproenkephalin in the nucleus accumbens shell. Reduced opioid neuropeptide transcription was accompanied by increased dynorphin and enkephalin peptide concentrations exclusively in 118G heroin subjects, suggesting that the peptide processing is associated with the OPRM1 genotype. Abnormal gene expression related to peptide convertase and ubiquitin/proteosome regulation was also evident in heroin users. Taken together, alterations in opioid neuropeptide systems might underlie enhanced opiate abuse vulnerability apparent in 118G individuals.
Collapse
Affiliation(s)
| | | | - Monika Cs Horváth
- Sections of *Psychiatry and
- Department of Pharmaceutical Biosciences, Division of Pharmacology, Uppsala University, S-751 24 Uppsala, Sweden; and
| | | | - Anna Gharibyan
- Alcohol and Drug Dependence Research, Department of Clinical Neuroscience, and
| | | | - Sadia Rahman
- Department of Pharmaceutical Biosciences, Division of Pharmacology, Uppsala University, S-751 24 Uppsala, Sweden; and
| | - Ingrid Nylander
- Department of Pharmaceutical Biosciences, Division of Pharmacology, Uppsala University, S-751 24 Uppsala, Sweden; and
| | - Georgy Bakalkin
- Alcohol and Drug Dependence Research, Department of Clinical Neuroscience, and
| | - Jovan Rajs
- Department of Forensic Medicine, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Eva Keller
- Department of Forensic Medicine, Semmelweis University, HU 1091, Budapest, Hungary
| | | |
Collapse
|
678
|
Abstract
PURPOSE OF REVIEW Alcohol dependence is a complex disorder with a well documented highly hereditary nature. This article reviews the recent advances in our understanding of the direct and indirect genetic influences on alcohol use and dependence. RECENT FINDINGS Recent findings can be summarized as follows: (a) twin studies have defined and estimated the risks of general and specific alcohol-related vulnerabilities. (b) Linkage studies have provided largely inconsistent findings, though several chromosomal regions have been implicated. (c) Quantitative trait loci analyses in animals have identified that the Mpdz gene predisposes to alcohol dependence and withdrawal. (d) Examination of family-based samples has identified several genes including GABRA2 and CHRM2 thought to be associated with alcohol dependence. SUMMARY Despite great advances in understanding of genetic vulnerability in alcohol use disorders, only two gene complexes, ADH and ALDH2, have been identified as having defined effects on alcohol use and liability to dependence in humans. New genes associated with increased risks for the disorder will certainly be added to this list in the near future. Neurobiological analyses of the effects of these genes will surely contribute to further understanding of the cause of alcohol dependence and the interindividual differences in risks.
Collapse
Affiliation(s)
- Susumu Higuchi
- National Hospital Organization, Kurihama Alcoholism Center, Yokosuka, Kanagawa, Japan.
| | | | | |
Collapse
|
679
|
Abstract
Twin, family, and adoption studies show that genetic factors play a significant role in vulnerability to becoming addicted to drugs. Through a combination of genetic approaches, the variant alleles responsible for interindividual differences in susceptibility to addiction are being uncovered.
Collapse
Affiliation(s)
- H M Lachman
- Albert Einstein College of Medicine, Departments of Medicine and Psychiatry, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| |
Collapse
|
680
|
McCarthy AM, Kleiber C. A conceptual model of factors influencing children's responses to a painful procedure when parents are distraction coaches. J Pediatr Nurs 2006; 21:88-98. [PMID: 16545669 DOI: 10.1016/j.pedn.2005.06.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The purpose of this article is to present a model of factors that may influence a child's response to a painful procedure when parents are distraction coaches during the procedure. Nonpharmacological interventions, in particular, distraction, and parents as coaches for their children during procedures are discussed. A conceptual model is presented that illustrates the multiple factors and their possible relationships. A selected review of studies is provided that supports the inclusion of these factors in the model. The model and literature review focus on three major areas: characteristics of the child, characteristics of the parent, and procedural variables. The model presented is currently being tested in a large multisite study on the use of distraction during intravenous line insertion.
Collapse
|
681
|
Rutter JL. Symbiotic relationship of pharmacogenetics and drugs of abuse. AAPS J 2006; 8:E174-84. [PMID: 16584126 PMCID: PMC2751437 DOI: 10.1208/aapsj080121] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2005] [Accepted: 01/18/2006] [Indexed: 02/03/2023] Open
Abstract
Pharmacogenetics/pharmacogenomics is the study of how genetic variation affects pharmacology, the use of drugs to treat disease. When drug responses are predicted in advance, it is easier to tailor medications to different diseases and individuals. Pharmacogenetics provides the tools required to identify genetic predictors of probable drug response, drug efficacy, and drug-induced adverse events-identifications that would ideally precede treatment decisions. Drug abuse and addiction genetic data have advanced the field of pharmacogenetics in general. Although major findings have emerged, pharmacotherapy remains hindered by issues such as adverse events, time lag to drug efficacy, and heterogeneity of the disorders being treated. The sequencing of the human genome and high-throughput technologies are enabling pharmacogenetics to have greater influence on treatment approaches. This review highlights key studies and identifies important genes in drug abuse pharmacogenetics that provide a basis for better diagnosis and treatment of drug abuse disorders.
Collapse
Affiliation(s)
- Joni L Rutter
- National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, 6001 Executive Boulevard, Bethesda, MD 20892, USA.
| |
Collapse
|
682
|
Zhang H, Luo X, Kranzler HR, Lappalainen J, Yang BZ, Krupitsky E, Zvartau E, Gelernter J. Association between two mu-opioid receptor gene (OPRM1) haplotype blocks and drug or alcohol dependence. Hum Mol Genet 2006; 15:807-19. [PMID: 16476706 PMCID: PMC3164878 DOI: 10.1093/hmg/ddl024] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
We examined 13 single nucleotide polymorphisms (SNPs) spanning the coding region of the mu-opioid receptor gene (OPRM1), among 382 European Americans (EAs) affected with substance dependence [alcohol dependence (AD) and/or drug dependence (DD)] and 338 EA healthy controls. These SNPs delineated two haplotype blocks. Genotype distributions for all SNPs were in Hardy-Weinberg equilibrium (HWE) in controls, but in cases, four SNPs in Block I and three SNPs in Block II showed deviation from HWE. Significant differences were found between cases and controls in allele and/or genotype frequencies for six SNPs in Block I and two SNPs in Block II. Association of SNP4 in Block I with DD (allele: P=0.004), SNP5 in Block I with AD and DD (allele: P< or =0.005 for both) and two SNPs in Block II with AD (SNP11 genotype: P=0.002; SNP12 genotype: P=0.001) were significant after correction for multiple testing. Frequency distributions of haplotypes (constructed by five tag SNPs) differed significantly for cases and controls (P<0.001 for both AD and DD). Logistic regression analyses confirmed the association between OPRM1 variants and substance dependence, when sex and age of subjects and alleles, genotypes, haplotypes or diplotypes of five tag SNPs were considered. Population structure analyses excluded population stratification artifact. Additional supporting evidence for association between OPRM1 and AD was obtained in a smaller Russian sample (247 cases and 100 controls). These findings suggest that OPRM1 intronic variants play a role in susceptibility to AD and DD in populations of European ancestry.
Collapse
Affiliation(s)
- Huiping Zhang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, Psychiatry 116A2, 950 Campbell Avenue, West Haven, CT 06516, USA
| | - Xingguang Luo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, Psychiatry 116A2, 950 Campbell Avenue, West Haven, CT 06516, USA
| | - Henry R. Kranzler
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Jaakko Lappalainen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, Psychiatry 116A2, 950 Campbell Avenue, West Haven, CT 06516, USA
| | - Bao-Zhu Yang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, Psychiatry 116A2, 950 Campbell Avenue, West Haven, CT 06516, USA
| | - Evgeny Krupitsky
- St Petersburg State Pavlov Medical University, St Petersburg, Russia
| | - Edwin Zvartau
- St Petersburg State Pavlov Medical University, St Petersburg, Russia
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, Psychiatry 116A2, 950 Campbell Avenue, West Haven, CT 06516, USA
| |
Collapse
|
683
|
Lötsch J, Geisslinger G. Current evidence for a genetic modulation of the response to analgesics. Pain 2006; 121:1-5. [PMID: 16472919 DOI: 10.1016/j.pain.2006.01.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2006] [Accepted: 01/12/2006] [Indexed: 10/25/2022]
Affiliation(s)
- Jörn Lötsch
- pharmazentrum frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe-University, Theodor Stern Kai 7, D-60590 Frankfurt, Germany.
| | | |
Collapse
|
684
|
Kreek MJ, Nielsen DA, Butelman ER, LaForge KS. Genetic influences on impulsivity, risk taking, stress responsivity and vulnerability to drug abuse and addiction. Nat Neurosci 2006; 8:1450-7. [PMID: 16251987 DOI: 10.1038/nn1583] [Citation(s) in RCA: 653] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Genetic variation may partially underlie complex personality and physiological traits--such as impulsivity, risk taking and stress responsivity--as well as a substantial proportion of vulnerability to addictive diseases. Furthermore, personality and physiological traits themselves may differentially affect the various stages of addiction, defined chronologically as initiation of drug use, regular drug use, addiction/dependence and potentially relapse. Here we focus on recent approaches to the study of genetic variation in these personality and physiological traits, and their influence on and interaction with addictive diseases.
Collapse
Affiliation(s)
- Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, New York, USA.
| | | | | | | |
Collapse
|
685
|
|
686
|
Bruehl S, Chung OY, Donahue BS, Burns JW. Anger Regulation Style, Postoperative Pain, and Relationship to the A118G Mu Opioid Receptor Gene Polymorphism: A Preliminary Study. J Behav Med 2006; 29:161-9. [PMID: 16400534 DOI: 10.1007/s10865-005-9030-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2005] [Accepted: 09/01/2005] [Indexed: 10/25/2022]
Abstract
Greater trait anger-out is associated with elevated pain responsiveness. Previous work suggests this effect may be mediated by deficient endogenous opioid analgesia, possibly reflecting diminished opioid receptor sensitivity. The A118G single nucleotide polymorphism (SNP) of the mu opioid receptor gene influences both opioid receptor sensitivity and clinical responsiveness to opioid analgesics. Therefore, this study tested whether this SNP either mediated or moderated the effects of anger-out on postsurgical pain outcomes. Forty-eight patients undergoing coronary artery bypass graft surgery provided genetic samples, and completed measures of anger-out and postsurgical pain. Postsurgical opioid analgesic use was also recorded. Anger-out was positively associated with postsurgical pain ratings (p < 0.05). Anger-out was not associated with A118G SNP status (p > 0.10), suggesting the latter is unlikely to mediate anger-out's pain-related effects. A significant anger-out x A118G interaction was observed on analgesic use (p < 0.05), due to a much stronger positive relationship between anger-out and analgesic demands in patient with the A118G SNP (b = 0.53) than those with the wild-type receptor (b = 0.07). These results suggest that the A118G SNP may moderate but not mediate the effects of anger-out on postoperative pain responses.
Collapse
Affiliation(s)
- Stephen Bruehl
- Department of Anesthesiology, Vanderbilt University School of Medicine, Tennessee, Nashville 37212, USA.
| | | | | | | |
Collapse
|
687
|
Abstract
It is generally assumed that combinations of polymorphic alleles of different genes contribute to polygenetic disorders. Variants of the opioid receptors are the obvious candidates underlying addiction. Most research has focused on the coding variation A118G of the mu opioid receptor (MOPr), which replaces asparagine at position 40 by aspartate (Asn40Asp). However, to date, no conclusive evidence exists regarding which physiological effects this mutation may cause. Other rare polymorphisms in the MOPr cause marked effects (e.g. impairment of G-protein coupling) but, due to their low frequency, their role in addiction is questionable. A large number of polymorphic sites have been found in the putative promoter region of the mu opioid receptor. These variations may alter the expression level of the receptors in neurones, but the functional relevance of promoter polymorphisms is hard to predict. In the delta opioid receptor, surprisingly few polymorphisms have been detected within the coding region, and a clear association with addiction has not been shown to date. The kappa opioid receptor contains mainly silent polymorphisms. Nevertheless, there are studies reporting positive associations of silent mutations in the three opioid receptors with drug addiction. The underlying mechanism remains unclear, but examples exist indicating that silent mutations affect mRNA stability. Taken together, the coding polymorphisms, which are rather frequent, reveal no convincing association. The vast number of non-coding, intronic or promoter polymorphisms in the opioid receptors may influence addictive behaviour, but these polymorphisms are far less studied, and their physiological significance remains to be demonstrated.
Collapse
MESH Headings
- Alleles
- Animals
- Humans
- Pharmacogenetics
- Polymorphism, Genetic
- 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
Collapse
Affiliation(s)
- Peter Mayer
- Bundesinstitut für Arzneimittel und Medizinprodukte, Bonn, Germany
| | | |
Collapse
|
688
|
Chong RY, Oswald L, Yang X, Uhart M, Lin PI, Wand GS. The mu-opioid receptor polymorphism A118G predicts cortisol responses to naloxone and stress. Neuropsychopharmacology 2006; 31:204-11. [PMID: 16123758 DOI: 10.1038/sj.npp.1300856] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A polymorphism in the mu-opioid receptor (MOR) (A118G) has been shown to increase beta-endorphin binding affinity, theoretically placing greater inhibitory tone on hypothalamic corticotropin-releasing hormone (CRH) neurons. We hypothesized that the minor allele (G) would predict cortisol responses to both pharmacological (naloxone) and psychological (stress) activation of the hypothalamic-pituitary-adrenal (HPA) axis. Healthy subjects (mean age 25.2 years, SD 9.2 years) completed a naloxone challenge (n=74) and/or the modified Trier Social Stress Test (TSST) (n=86). For the naloxone challenge, two baseline blood samples were obtained. Then, five increasing doses of i.v. naloxone were administered at 30-min intervals and 12 additional blood samples were collected at 15-min intervals. The TSST consisted of 5-min of public speaking and 5-min of mental arithmetic exercises. Three baseline and five post-TSST blood samples were drawn. Both the naloxone and TSST groups had significant adrenocorticotropin (ACTH) and cortisol responses to their respective challenges (P<0.001). There were no differences in baseline ACTH, baseline cortisol, or ACTH response by genotype in either the naloxone or the TSST group. Among subjects expressing a G allele, there was a higher cortisol response to naloxone (P=0.046), but a lower cortisol response to the TSST (P=0.044). In conclusion, the minor allele (G) was associated with a robust cortisol response to naloxone blockade, but a blunted response to psychosocial stress. We speculate that increased opioid avidity of the minor allele receptor contributes to the differential response to naloxone vs stress.
Collapse
Affiliation(s)
- Rachel Y Chong
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
689
|
Gallagher CJ, Gordon CJ, Langefeld CD, Mychaleckyj JC, Freedman BI, Rich SS, Bowden DW, Sale MM. Association of the mu-opioid receptor gene with type 2 diabetes mellitus in an African American population. Mol Genet Metab 2006; 87:54-60. [PMID: 16140553 DOI: 10.1016/j.ymgme.2005.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2005] [Accepted: 07/19/2005] [Indexed: 11/24/2022]
Abstract
African Americans (AA) are at increased risk for developing type 2 diabetes mellitus (T2DM) relative to European Americans. We previously detected linkage of T2DM to 6q24-q27 (LOD 2.26) at 163.5 cM, closest to marker D6S1035, in a genome-wide scan of AA families. The mu-opioid receptor gene (OPRM1) is located within the LOD-1 support interval of this linkage peak. OPRM1 is an attractive positional candidate gene for T2DM susceptibility since agonists of OPRM1 affect glucose-induced insulin release and OPRM1 knockout mice have a more rapid induction of insulin resistance than wild-type. Twenty-two SNPs in this gene, at an average spacing of 3.9 kb, were genotyped in 380 AA T2DM cases and 276 AA controls. In single SNP association analyses, rs648007 demonstrated significant evidence of association with T2DM (P=0.013). Four blocks of high linkage disequilibrium were detected across the OPRM1 gene. Association analyses of haplotypes in each of these blocks revealed two haplotype blocks with significant overall P values (P=0.007 and 0.046). Significant, but rare, risk and protective haplotypes were identified as driving these associations with T2DM (P=0.034-0.047). These associations suggest that the OPRM1 gene plays a role in T2DM susceptibility in African Americans.
Collapse
Affiliation(s)
- Carla J Gallagher
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | | | | | | | |
Collapse
|
690
|
Abstract
Smoking is a complex behavior influenced by environ-mental and genetic factors. A number of twin studies demonstrate genetic influences on multiple aspects of smoking including initiation, dependence, continuation, amount consumed, and cessation. It is likely that this genetic risk involves a complex interaction between multiple genes in different biologic pathways. These may include genes involved in the general aspect of dependence (eg, genes within the dopaminergic system) and those more specific to nicotine dependence (eg, genes encoding nicotinic acetylcholine receptors and nicotine-metabolizing enzymes). Genome-wide linkage analyses and gene association studies provide approaches for identifying these genes. This review provides a brief highlight of data from some of the candidate gene association studies and discusses some of the discrepancies found in the literature. In addition, the authors have reviewed some recent studies examining how genetic variations in drug-metabolizing enzymes and drug targets can influence the response to treatment with drugs such as bupropion and nicotine.These latter studies provide the potential for maximizing treatment efficacy by individualizing the type, dosage, and duration of the treatment based on an individual's genotype/phenotype. Significant advances have been made in understanding the genetic contribution to smoking behaviors. These and future studies will provide potential directions for novel prevention and treatment strategies.
Collapse
Affiliation(s)
- Nael Al Koudsi
- Department of Pharmacology, Centre for Addiction and Mental Health, University of Toronto, Canada
| | | |
Collapse
|
691
|
Tang CM, Insel PA. Genetic variation in G-protein-coupled receptors – consequences for G-protein-coupled receptors as drug targets. Expert Opin Ther Targets 2005; 9:1247-65. [PMID: 16300474 DOI: 10.1517/14728222.9.6.1247] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
G-protein-coupled receptors (GPCRs), including 'orphan' GPCRs whose natural ligands are unknown, comprise the largest membrane receptor superfamily and are the most commonly used therapeutic targets. GPCR genetic loci harbour numerous variants, such as DNA insertions or deletions and single nucleotide polymorphisms that alter GPCR expression and function, thereby contributing to inter-individual differences in disease susceptibility/progression and drug responses. In this article, the authors review examples of GPCR genetic variants that influence transcription, translation, receptor folding and expression on cell surface (by affecting receptor trafficking, dimerisation, desensitisation/downregulation), or perturb receptor function (by altering ligand binding, G-protein coupling and receptor constitutive activity). In spite of such effects, assessment for genetic variants is not currently applied to the drug development and approval process or in the clinical use of GPCR drugs. Further insights will, the authors believe, alter drug discovery/development, therapeutics and likely provide new GPCR drug targets.
Collapse
Affiliation(s)
- Chih-Min Tang
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093-0636, USA
| | | |
Collapse
|
692
|
Funck-Brentano C, Boëlle PY, Verstuyft C, Bornert C, Becquemont L, Poirier JM. Measurement of CYP2D6 and CYP3A4 activity in vivo with dextromethorphan: sources of variability and predictors of adverse effects in 419 healthy subjects. Eur J Clin Pharmacol 2005; 61:821-9. [PMID: 16315033 DOI: 10.1007/s00228-005-0051-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2005] [Accepted: 10/10/2005] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Dextromethorphan (DEM) shares part of the adverse event profile of opioids and is widely used as a probe drug for CYP2D6 phenotyping and for the assessment of CYP2D6 activity. It has also been used to assess CYP3A4 activity. This study examined the influence of anthropometric variables, oral contraceptives, smoking habits, mu-opioid receptor and MDR1 genetic polymorphisms and components of the DEM ratios on the variability of CYP2D6 and CYP3A4 metabolic ratios and on the occurrence of adverse events following DEM administration. METHODS This was a retrospective analysis of a database in 419 healthy subjects. CYP2D6 and CYP3A4 metabolic ratios were measured as the log of the ratios of the amount of DEM to the amount of dextrorphan (DOR) and of the amount of DEM to the amount of 3-methoxy-morphinan (MET) excreted in urine during a 12-h time period, respectively, following the oral administration of 80 mg of dextromethorphan hydrobromide. Logistic regression was performed to examine the factors associated with changes in metabolic ratios and with the occurrence of adverse events. RESULTS The CYP2D6 metabolic ratio allowed identification of extensive and poor metabolizers of DEM. The CYP2D6 and CYP3A4 metabolic ratios were not strictly independent one from each other. Based on multivariate analysis, the CYP2D6 metabolic ratio was a stronger independent predictor of adverse events (p<0.0001) than the CYP2D6 phenotype (p=0.05). Anthropometric variables, oral contraceptives, smoking habits, mu-opioid receptor and MDR1 genetic polymorphisms did not significantly contribute to changes in metabolic ratios or to the occurrence of adverse events. CONCLUSIONS Dextromethorphan can be used for CYP2D6 phenotyping, but the CYP2D6 and CYP3A4 metabolic ratios are not strictly independent one from each other. The CYP2D6 metabolic ratio predicts adverse events to DEM as does CYP2D6 phenotype, and extensive metabolizer subjects are not protected against adverse events.
Collapse
Affiliation(s)
- Christian Funck-Brentano
- INSERM--AP-HP, Clinical Investigation Center, CIC-9304, and Department of Pharmacology, Pierre et Marie Curie University, 75012 Paris, France.
| | | | | | | | | | | |
Collapse
|
693
|
Yuferov V, Butelman ER, Kreek MJ. Biological clock: biological clocks may modulate drug addiction. Eur J Hum Genet 2005; 13:1101-3. [PMID: 16094306 DOI: 10.1038/sj.ejhg.5201483] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
694
|
Edenberg HJ, Kranzler HR. The contribution of genetics to addiction therapy approaches. Pharmacol Ther 2005; 108:86-93. [PMID: 16026844 DOI: 10.1016/j.pharmthera.2005.06.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2005] [Accepted: 06/16/2005] [Indexed: 11/25/2022]
Abstract
Addictions, including alcohol dependence, which is the focus of this article, are complex genetic diseases. Recently, several individual genes that contribute to the risk for alcohol dependence have been identified, and more are expected to be in the near future. Among these are genes encoding alcohol and aldehyde dehydrogenases and GABA(A) receptor subunits. These reveal pathways of vulnerability and provide targets for rational drug design. It is likely that response to particular therapies is also a complex trait influenced by genetics, but studies to explore this are just beginning. We discuss some studies on bromocriptine, naltrexone, and serotonergic agents. Adding a genetic component to treatment trials could greatly help to understand the biological basis of variations in the efficacy of therapies and, in the future, could lead to individualized choices of therapy.
Collapse
Affiliation(s)
- Howard J Edenberg
- Indiana University School of Medicine, 635 Barnhill Drive, MS4063, Indianapolis, IN 46202-5122, USA.
| | | |
Collapse
|
695
|
Landau R. Pharmacogenetics: implications for obstetric anesthesia. Int J Obstet Anesth 2005; 14:316-23. [PMID: 16143506 DOI: 10.1016/j.ijoa.2005.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2005] [Accepted: 03/01/2005] [Indexed: 11/22/2022]
Affiliation(s)
- Ruth Landau
- Service d'Anesthésiologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland.
| |
Collapse
|
696
|
Zhang Y, Wang D, Johnson AD, Papp AC, Sadée W. Allelic Expression Imbalance of Human mu Opioid Receptor (OPRM1) Caused by Variant A118G. J Biol Chem 2005; 280:32618-24. [PMID: 16046395 DOI: 10.1074/jbc.m504942200] [Citation(s) in RCA: 444] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
As a primary target for opioid drugs and peptides, the mu opioid receptor (OPRM1) plays a key role in pain perception and addiction. Genetic variants of OPRM1 have been implicated in predisposition to drug addiction, in particular the single nucleotide polymorphism A118G, leading to an N40D substitution, with an allele frequency of 10-32%, and uncertain functions. We have measured allele-specific mRNA expression of OPRM1 in human autopsy brain tissues, using A118G as a marker. In 8 heterozygous samples measured, the A118 mRNA allele was 1.5-2.5-fold more abundant than the G118 allele. Transfection into Chinese hamster ovary cells of a cDNA representing only the coding region of OPRM1, carrying adenosine, guanosine, cytidine, and thymidine in position 118, resulted in 1.5-fold lower mRNA levels only for OPRM1-G118, and more than 10-fold lower OPRM1 protein levels, measured by Western blotting and receptor binding assay. After transfection and inhibition of transcription with actinomycin D, analysis of mRNA turnover failed to reveal differences in mRNA stability between A118 and G118 alleles, indicating a defect in transcription or mRNA maturation. These results indicate that OPRM1-G118 is a functional variant with deleterious effects on both mRNA and protein yield. Clarifying the functional relevance of polymorphisms associated with susceptibility to a complex disorder such as drug addiction provides a foundation for clinical association studies.
Collapse
MESH Headings
- Alleles
- Allelic Imbalance
- Animals
- Blotting, Western
- Brain/metabolism
- CHO Cells
- Cell Line
- Cricetinae
- DNA/chemistry
- DNA, Complementary/metabolism
- Dactinomycin/pharmacology
- Gene Frequency
- Genetic Predisposition to Disease
- Genetic Variation
- Genotype
- Heterozygote
- Humans
- Mutagenesis
- Mutagenesis, Site-Directed
- Nucleic Acid Conformation
- Nucleic Acid Synthesis Inhibitors/pharmacology
- Plasmids/metabolism
- Polymorphism, Genetic
- Polymorphism, Single Nucleotide
- RNA, Messenger/metabolism
- Receptors, Opioid, mu/biosynthesis
- Receptors, Opioid, mu/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Substance-Related Disorders
- Time Factors
- Transcription, Genetic
- Transfection
Collapse
Affiliation(s)
- Ying Zhang
- Program in Pharmacogenomics, Department of Pharmacology, College of Medicine and Public Health, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | |
Collapse
|
697
|
Lötsch J, Freynhagen R, Geisslinger G. Sind μ-Opioidrezeptorpolymorphismen wichtig für die Opioidtherapie? Schmerz 2005; 19:378-82, 384-5. [PMID: 16133302 DOI: 10.1007/s00482-005-0423-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Polymorphisms in the mu-opioid receptor gene may potentially alter the clinical effects of opioid analgesics. A common mu-opioid receptor polymorphism occurring at an allelic frequency of 12% decreases the potency of opioid analgesics in humans. Interestingly, in carriers of this mutation, it appears to be possible to reach analgesia by increasing the opioid dose but side effects appear to occur less often despite the higher opioid dose. This suggests a broadened therapeutic range of the opioids. Other mutations of the mu-opioid receptor, for example three mutations within the third intracellular loop of the receptor, impair receptor signaling, but they are too rare to greatly affect pain therapy or have not yet been investigated in the context of pain therapy.
Collapse
Affiliation(s)
- J Lötsch
- Pharmazentrum Frankfurt/ZAFES, Institut für Klinische Pharmakologie, Johann-Wolfgang-Goethe-Universität, Frankfurt am Main.
| | | | | |
Collapse
|
698
|
Ide S, Han W, Kasai S, Hata H, Sora I, Ikeda K. Characterization of the 3' untranslated region of the human mu-opioid receptor (MOR-1) mRNA. Gene 2005; 364:139-45. [PMID: 16122888 DOI: 10.1016/j.gene.2005.05.040] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2005] [Revised: 04/07/2005] [Accepted: 05/30/2005] [Indexed: 11/30/2022]
Abstract
The mu-opioid receptor (MOR) plays a mandatory role in the action of most opioid drugs, such as morphine, fentanyl, and heroin. It has been revealed that a deficiency in the MOR gene (Oprm1) or a difference in the 3' noncoding region of the gene markedly affects the sensitivity of mice to opioids. As the 3' noncoding region of the human OPRM1 gene had not yet been characterized, in the present study we conducted 3'-rapid amplification of cDNA ends (3'RACE)-PCR and identified the 3' end of the human MOR-1 mRNA, the most abundant transcript among OPRM1 gene transcripts. The poly(A) signal was located at 13612-13617 nucleotides downstream from the stop codon in the OPRM1 gene. Reverse transcription PCR analyses showed that the region from the stop codon to the poly(A) signal was transcribed. In the 3'UTR, we identified 33 AU-rich regions and more than 300 putative transcription factor-binding sites. Furthermore, we compared the 3' noncoding regions of the human and mouse OPRM1/Oprm1 genes and found apparent homology. In Northern blotting with mouse brain mRNAs, a same-size band was detected by a probe for the MOR-1 coding region and by a probe for a mouse genome region corresponding to the human MOR-1 3'UTR. Since 3'UTRs affect gene expression, the present characterization of the 3' noncoding region in the human OPRM1 gene should lead to a better understanding of the mechanisms underlying OPRM1 gene regulation and individual differences in sensitivity to opioids.
Collapse
Affiliation(s)
- Soichiro Ide
- Division of Psychobiology, Tokyo Institute of Psychiatry, 2-1-8 Kamikitazawa, Setagaya-ku, Tokyo 156-8585, Japan
| | | | | | | | | | | |
Collapse
|
699
|
Ross JR, Rutter D, Welsh K, Joel SP, Goller K, Wells AU, Du Bois R, Riley J. Clinical response to morphine in cancer patients and genetic variation in candidate genes. THE PHARMACOGENOMICS JOURNAL 2005; 5:324-36. [PMID: 16103897 DOI: 10.1038/sj.tpj.6500327] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Morphine is the analgesic of choice for moderate to severe cancer pain; however, 10-30% of patients do not tolerate morphine. This study evaluated genetic variation in the mu-opioid receptor, betaarrestin2, stat6 and uridine diphosphate-glucuronysltransferase 2B7 (UGT2B7) genes, in patients who responded to morphine vs those who were switched to alternative opioids. We prospectively recruited and genotyped 162 Caucasian patients (117 controls, 39 switchers). Switchers, were more likely to carry the common allele at 1182 G/A, 5864 G/A, 8622T/C and 11143 G/A in the betaarrestin2 gene (P = 0.021, 0.043, 0.013, 0.043, respectively). Switchers had increased carriage of the T allele (-1714 C/T) and a significant difference in the allelic frequency at 9065 C/T (chi(2) = 3.86, P = 0.049) in the stat6 gene. No differences were seen in genotype or allele frequencies of SNPs in the mu-opioid receptor gene or UGT2B7 gene. This study presents novel data suggesting that variation in genes involved in mu-opioid receptor signalling influence clinical response to morphine.
Collapse
Affiliation(s)
- J R Ross
- Department of Clinical Genomics, Imperial College, London, UK.
| | | | | | | | | | | | | | | |
Collapse
|
700
|
Szumlinski KK, Lominac KD, Frys KA, Middaugh LD. Genetic variation in heroin-induced changes in behaviour: effects of B6 strain dose on conditioned reward and locomotor sensitization in 129-B6 hybrid mice. GENES BRAIN AND BEHAVIOR 2005; 4:324-36. [PMID: 16011579 DOI: 10.1111/j.1601-183x.2004.00111.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Substantial interindividual variability exists in the propensity to develop opiate addiction. Genetic variation in opiate reward may contribute to this variability. A large body of evidence indicates genetic variation in mice for several effects of opiate drugs. The present study examined heroin-induced place conditioning and locomotor sensitization in the two strains of mice employed most frequently in the generation of transgenic animals, C57BL/6J (B6) and 129X1/sVJ (129), as well as in groups of B6-129 hybrid mice, differing in their amount of B6 genetic background. Four pairings of 100 microg/kg of heroin elicited robust place conditioning and locomotor sensitization in B6 controls and in N(10) congenic B6-129 hybrid mice. In comparison, the identical treatment produced no locomotor sensitization and induced place aversion in 129 controls. No heroin-induced changes in the behaviour of N(3) congenic B6-129 hybrid mice or F5-8 non-congenic B6-129 hybrid mice were observed. The expression of place conditioning was not facilitated in any group by the administration of a heroin-priming injection prior to testing. These data indicate that genetic variation exists in mice for the rewarding and locomotor-sensitizing effects of heroin and that the capacity of heroin to induce conditioned reward and locomotor sensitization can be modulated in a B6 strain dose-dependent manner in B6-129 hybrid mice. Thus, strain differences in heroin responsiveness should be considered when examining transgenic lines on B6-129 backgrounds for opiate-induced changes in behaviour that may be relevant for addiction.
Collapse
Affiliation(s)
- K K Szumlinski
- Department of Physiology and Neuroscience, and Department of Psychiatry and Behavioral Sciences, Center for Drug and Alcohol Programs, Medical University of South Carolina, Charleston, SC, USA.
| | | | | | | |
Collapse
|