1
|
Bean BDM, Mulvihill CJ, Garge RK, Boutz DR, Rousseau O, Floyd BM, Cheney W, Gardner EC, Ellington AD, Marcotte EM, Gollihar JD, Whiteway M, Martin VJJ. Functional expression of opioid receptors and other human GPCRs in yeast engineered to produce human sterols. Nat Commun 2022; 13:2882. [PMID: 35610225 PMCID: PMC9130329 DOI: 10.1038/s41467-022-30570-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
The yeast Saccharomyces cerevisiae is powerful for studying human G protein-coupled receptors as they can be coupled to its mating pathway. However, some receptors, including the mu opioid receptor, are non-functional, which may be due to the presence of the fungal sterol ergosterol instead of cholesterol. Here we engineer yeast to produce cholesterol and introduce diverse mu, delta, and kappa opioid receptors to create sensitive opioid biosensors that recapitulate agonist binding profiles and antagonist inhibition. Additionally, human mu opioid receptor variants, including those with clinical relevance, largely display expected phenotypes. By testing mu opioid receptor-based biosensors with systematically adjusted cholesterol biosynthetic intermediates, we relate sterol profiles to biosensor sensitivity. Finally, we apply sterol-modified backgrounds to other human receptors revealing sterol influence in SSTR5, 5-HTR4, FPR1, and NPY1R signaling. This work provides a platform for generating human G protein-coupled receptor-based biosensors, facilitating receptor deorphanization and high-throughput screening of receptors and effectors.
Collapse
Affiliation(s)
- Björn D M Bean
- Department of Biology, Centre for Applied Synthetic Biology, Concordia University, Montréal, QC, H4B1R6, Canada
| | - Colleen J Mulvihill
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Riddhiman K Garge
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Daniel R Boutz
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
- DEVCOM Army Research Laboratory-South, Austin, 78712, TX, USA
| | - Olivier Rousseau
- Department of Biology, Centre for Applied Synthetic Biology, Concordia University, Montréal, QC, H4B1R6, Canada
| | - Brendan M Floyd
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - William Cheney
- Department of Biology, Centre for Applied Synthetic Biology, Concordia University, Montréal, QC, H4B1R6, Canada
| | - Elizabeth C Gardner
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew D Ellington
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Edward M Marcotte
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Jimmy D Gollihar
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA.
- DEVCOM Army Research Laboratory-South, Austin, 78712, TX, USA.
- Center for Molecular and Translational Human Infectious Diseases Research, Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA.
| | - Malcolm Whiteway
- Department of Biology, Centre for Applied Synthetic Biology, Concordia University, Montréal, QC, H4B1R6, Canada
| | - Vincent J J Martin
- Department of Biology, Centre for Applied Synthetic Biology, Concordia University, Montréal, QC, H4B1R6, Canada.
| |
Collapse
|
2
|
Bedoya F, Meneu JC, Macías MI, Moreno A, Enríquez-de-Salamanca R, Gonzalez EM, Vegh I. Mutation in CNR1 Gene and VEGF Expression in Esophageal Cancer. TUMORI JOURNAL 2018; 95:68-75. [DOI: 10.1177/030089160909500112] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims and background Cannabinoid receptors have an impact on gastrointestinal function, but it remains unknown whether mutations may affect tumor susceptibility in patients with esophageal carcinoma. The aim of this study was to determine mutation in the cannabinoid receptor-1 (CNR1) gene and its relation to vascular endothelial growth factor (VEGF) expression as an angiogenic and poor prognostic factor. Methods 179 esophageal tissue samples from 69 patients (29 with esophageal cancer and 40 controls) were studied. CNR1 gene mutation (1359 G → A in codon 453) was detected with PCR, using the MspI restriction enzyme. VEGF was determined by immunoassay. Results Genotyping in control patients’ samples revealed that 24/40 were G/G wild type and 16/40 were G/A; no samples were A/A. Of the 139 tissue samples from the 29 esophageal cancer patients, 15 were G/G homozygous, 85 G/A heterozygous, 11 had an A/A genotype and 28 were without amplification. In the normal tissue adjacent to tumor, some mutations were observed. The overall survival time was reduced in patients with the A/A type in all their 5 samples, in comparison to G/G type (P = 0.04, chi-square: 4.26). VEGF expression was higher in tumor than nontumor areas (P <0.025). VEGF expression was not correlated with survival time. Conclusions Our preliminary findings in esophageal tissue showed a high frequency of G → A mutation in the CNR1 gene. No correlation between VEGF expression and gene receptor mutation was found. Patients with mutation in all their samples had a reduced survival time.
Collapse
Affiliation(s)
- Fernando Bedoya
- Research Center, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Juan C Meneu
- Department of Surgery, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Almudena Moreno
- Department of Surgery, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | - Irene Vegh
- Research Center, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
3
|
Association of the OPRM1 and COMT genes’ polymorphisms with the efficacy of morphine in Tunisian cancer patients: Impact of the high genetic heterogeneity in Tunisia? Therapie 2016; 71:507-513. [DOI: 10.1016/j.therap.2016.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 04/04/2016] [Indexed: 01/21/2023]
|
4
|
Cieślińska A, Sienkiewicz-Szłapka E, Wasilewska J, Fiedorowicz E, Chwała B, Moszyńska-Dumara M, Cieśliński T, Bukało M, Kostyra E. Influence of candidate polymorphisms on the dipeptidyl peptidase IV and μ-opioid receptor genes expression in aspect of the β-casomorphin-7 modulation functions in autism. Peptides 2015; 65:6-11. [PMID: 25625371 DOI: 10.1016/j.peptides.2014.11.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/07/2014] [Accepted: 11/08/2014] [Indexed: 02/08/2023]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder with population prevalence of approximately 60-70 per 10,000. Data shows that both opioid system function enhancement and opiate administration can result in autistic-like symptoms. Cow milk opioid peptides, including β-casomorphin-7 (BCM7, Tyr-Pro-Phe-Pro-Gly-Pro-Ile), affect the μ-opioid receptor (MOR) and are subjected to degradation resulting from the proline dipeptidyl peptidase IV (DPPIV, EC 3.4.14.5) enzyme activity. The presence of MOR and DPPIV activity are crucial factors determining biological activity of BCM7 in the human body. Our study examined the effect of β-casomorphin-7 on the MOR and DPPIV genes expression according to specific point mutations in these genes. In addition, we investigated frequency of A118G SNP in the MOR gene and rs7608798 of the DPPIV (A/G) gene in healthy and autistic children. Our research indicated correlation in DPPIV gene expression under the influence of BCM7 and hydrolyzed milk between healthy and ASD-affected children with genotype GG (P<0.0001). We also observed increased MOR gene expression in healthy children with genotype AG at polymorphic site A118G under influence of BCM7 and hydrolyzed milk. The G allele frequency was 0.09 in MOR gene and 0.68 in the DPPIV gene. But our results suggest no association between presence of the alleles G and A at position rs7608798 in DPPIV gene nor alleles A and G at position A118G of the MOR and increased incidence of ASD. Our studies emphasize the compulsion for genetic analysis in correlation with genetic factors affecting development and enhancement of autism symptoms.
Collapse
Affiliation(s)
- Anna Cieślińska
- Faculty of Biology and Biotechnology, University of Warmia and Mazury, Oczapowskiego 1A Street, 10-19 Olsztyn, Poland
| | - Edyta Sienkiewicz-Szłapka
- Faculty of Biology and Biotechnology, University of Warmia and Mazury, Oczapowskiego 1A Street, 10-19 Olsztyn, Poland
| | - Jolanta Wasilewska
- Department of Paediatrics, Gastroenterology and Allergology, Medical University of Białystok, Poland
| | - Ewa Fiedorowicz
- Faculty of Biology and Biotechnology, University of Warmia and Mazury, Oczapowskiego 1A Street, 10-19 Olsztyn, Poland
| | - Barbara Chwała
- Regional Children's Hospital in Olsztyn, Zolnierska 18 A Street, 10-561 Olsztyn, Poland
| | - Małgorzata Moszyńska-Dumara
- Center for Diagnosis, Treatment and Therapy of Autism at the Regional Children's Hospital in Olsztyn, Zolnierska 18 A Street, 10-561 Olsztyn, Poland
| | - Tomasz Cieśliński
- Center for Diagnosis, Treatment and Therapy of Autism at the Regional Children's Hospital in Olsztyn, Zolnierska 18 A Street, 10-561 Olsztyn, Poland
| | - Marta Bukało
- Faculty of Biology and Biotechnology, University of Warmia and Mazury, Oczapowskiego 1A Street, 10-19 Olsztyn, Poland
| | - Elżbieta Kostyra
- Faculty of Biology and Biotechnology, University of Warmia and Mazury, Oczapowskiego 1A Street, 10-19 Olsztyn, Poland.
| |
Collapse
|
5
|
Sun Y, Meng S, Li J, Shi J, Lu L. Advances in genetic studies of substance abuse in China. SHANGHAI ARCHIVES OF PSYCHIATRY 2014; 25:199-211. [PMID: 24991158 PMCID: PMC4054556 DOI: 10.3969/j.issn.1002-0829.2013.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Summary The importance of genetic factors in substance addiction has long been established. The rationale for this work is that understanding of the function of addiction genes and delineation of the key molecular pathways of these genes would enhance the development of novel therapeutic targets and biomarkers that could be used in the prevention and management of substance abuse. Over the past few years, there has been a substantial increase in the number of genetic studies conducted on addiction in China; these studies have primarily focused on heroin, alcohol, and nicotine dependence. Most studies of candidate genes have concentrated on the dopamine, opioid, and serotonin systems. A number of genes associated with substance abuse in Caucasians are also risk factors in Chinese, but several novel genes and genetic risk factors associated with substance abuse in Chinese subjects have also been identified. This paper reviews the genetic studies of substance abuse performed by Chinese researchers. Genotypes and alleles related to addictive behavior in Chinese individuals are discussed and the contributions of Chinese researchers to the international corpus of knowledge about the genetic understanding of substance abuse are described.
Collapse
Affiliation(s)
- Yan Sun
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Shiqiu Meng
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Jiali Li
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Jie Shi
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Lin Lu
- Institute of Mental Health, Peking University, Beijing, China
| |
Collapse
|
6
|
Mamie C, Rebsamen MC, Morris MA, Morabia A. First Evidence of a Polygenic Susceptibility to Pain in a Pediatric Cohort. Anesth Analg 2013; 116:170-7. [DOI: 10.1213/ane.0b013e31826f0637] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
7
|
Huehne K, Leis S, Muenster T, Wehrfritz A, Winter S, Maihöfner C, Foertsch T, Croner R, Reis A, Winterpacht A, Rautenstrauss B. High post surgical opioid requirements in Crohn's disease are not due to a general change in pain sensitivity. Eur J Pain 2012; 13:1036-42. [DOI: 10.1016/j.ejpain.2008.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Revised: 12/01/2008] [Accepted: 12/14/2008] [Indexed: 11/30/2022]
|
8
|
What can allostasis tell us about anabolic-androgenic steroid addiction? Dev Psychopathol 2011; 23:907-19. [PMID: 21756441 DOI: 10.1017/s0954579411000393] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Anabolic-androgenic steroids (AASs) are synthetic hormones used by individuals who want to look better or perform better in athletics and at the gym. Their use raises an interesting paradox in which drug use is associated with a number of health benefits, but also the possibility of negative health consequences. Existing models of AAS addiction follow the traditional framework of drug abuse and dependence, which suggest that harmful use occurs as a result of the drug's ability to hijack the motivation-reward system. However, AASs, unlike typical drugs of abuse, are not used for acute intoxication effects or euphoria. Rather, AASs are used to affect the body through changes to the musculoskeletal system and the hypothalamic-pituitary-gonadal axis as opposed to stimulating the reward system. We offer an allostatic model of AAS addiction to resolve this inconsistency between traditional drug addiction and AAS addiction. This allostatic framework provides a way to (a) incorporate exercise into AAS misuse, (b) identify where AAS use transitions from recreational use into a drug problem, and (c) describe individual differences in vulnerability or resilience to AASs. Implications for this model of AAS addiction are discussed.
Collapse
|
9
|
Khokhar JY, Ferguson CS, Zhu AZX, Tyndale RF. Pharmacogenetics of drug dependence: role of gene variations in susceptibility and treatment. Annu Rev Pharmacol Toxicol 2010; 50:39-61. [PMID: 20055697 DOI: 10.1146/annurev.pharmtox.010909.105826] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Drug dependency is a highly prevalent mental health disorder that imposes a significant burden on those directly affected, health care systems, and society in general. There is substantial heritability in the susceptibility to drug addiction, which indicates that there are genetic risk factors. Variation in the human genome is abundant and can directly affect drug dependency phenotypes, for example, by altering the function of a gene product or by altering gene expression. Pharmacogenetic studies can assess the effects of genetic variation on the risk for a particular phenotype (e.g., being an alcoholic). In addition, pharmacogenetic variability in treatment efficacy and adverse reactions can be investigated to identify particular genetic variants associated with altered responses. This review highlights examples of genetic variations that are important in the development and maintenance of specific drug dependencies as well as those that affect the response to treatment.
Collapse
Affiliation(s)
- Jibran Y Khokhar
- The Center for Addiction and Mental Health and the Department of Pharmacology and Toxicology, University of Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
10
|
|
11
|
Leskelä TT, Markkanen PMH, Alahuhta IA, Tuusa JT, Petäjä-Repo UE. Phe27Cys Polymorphism Alters the Maturation and Subcellular Localization of the Human δ Opioid Receptor. Traffic 2008; 10:116-29. [DOI: 10.1111/j.1600-0854.2008.00846.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
12
|
Abrahao KP, Quadros IM, Souza-Formigoni MLO. Morphine attenuates the expression of sensitization to ethanol, but opioid antagonists do not. Neuroscience 2008; 156:857-64. [PMID: 18804151 DOI: 10.1016/j.neuroscience.2008.08.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 08/08/2008] [Accepted: 08/09/2008] [Indexed: 01/30/2023]
Abstract
Behavioral sensitization to ethanol is characterized by an increased locomotor activity after repeated exposure. A great variability exists among species and strains in the development of sensitization. There is a growing amount of evidence to indicate that the opioid system is involved in alcoholism; it is possible, therefore, that this system also modulates the sensitization to ethanol. In this study we evaluated the role of the opioid system in determining the variability of the sensitized response to ethanol. Mice received repeated administrations of ethanol (2.2 g/kg) or saline every other day for 10 days. According to their locomotor response on the last day of treatment, ethanol-treated animals were classified into two groups: sensitized or non-sensitized mice. After the treatment, mice were submitted to four challenges 48 h apart. In experiments 1 and 2, mice were challenged, respectively, with i.p. administration of opioid antagonists (naloxone or naltrexone) or an opioid agonist (morphine), followed immediately by 2.2 g/kg ethanol. In experiment 3, animals received morphine by i.c.v., followed by 2.2 g/kg of ethanol (i.p.). Pretreatment with opioid antagonists (naloxone or naltrexone) did not block the expression of ethanol sensitization; however pretreatment with morphine attenuated the increased locomotor activity after ethanol administration in sensitized mice. In experiment 4, after the ethanol or saline treatment, mice brains were processed and brain mu opioid binding was assessed by autoradiography using [3H]D-Ala2,N-mePhe4, Gly-ol5-enkephalin ([3H]DAMGO). No differences were seen between any of the groups of mice, so the agonist effect is not likely to be mediated by differences in binding to mu opioid receptors.
Collapse
Affiliation(s)
- K P Abrahao
- Departamento de Psicobiologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | | | | |
Collapse
|
13
|
Human expression variation in the mu-opioid receptor is paralleled in rhesus macaque. Behav Genet 2008; 38:390-5. [PMID: 18379868 DOI: 10.1007/s10519-008-9207-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Accepted: 03/19/2008] [Indexed: 10/22/2022]
Abstract
The mu-opioid receptor is a key component in many neurobiological systems including those affecting perceptions of pain and pleasure. In humans and non-human primate model systems, genetic variation in the receptor has been associated with numerous behavioral and physiological traits. In humans, polymorphisms have been identified which affect not only the biochemical function of the receptor, but also expression level. Existing rhesus macaque variation parallels the functional protein changes seen in human, but it remains unknown if expression level differences or concomitant protein changes may also exist. Here we perform a comprehensive survey of naturally occurring polymorphisms in Indian-origin rhesus macaques and identify three 5' UTR haplotypes with effects on expression level. These expression level effects are in linkage disequilibrium with the previously identified rhesus coding polymorphism C77G. The C77G polymorphism in rhesus parallels the functional effects of the A118G polymorphism in humans and expression level differences occur within both species. Together, the functional variations reported here have implications for future studies seeking to model the opioid system and its associated phenotypes in rhesus macaques.
Collapse
|
14
|
Nielsen DA, Ji F, Yuferov V, Ho A, Chen A, Levran O, Ott J, Kreek MJ. Genotype patterns that contribute to increased risk for or protection from developing heroin addiction. Mol Psychiatry 2008; 13:417-28. [PMID: 18195715 PMCID: PMC3810149 DOI: 10.1038/sj.mp.4002147] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Accepted: 12/06/2007] [Indexed: 11/09/2022]
Abstract
A genome-wide association study was conducted using microarray technology to identify genes that may be associated with the vulnerability to develop heroin addiction, using DNA from 104 individual former severe heroin addicts (meeting Federal criteria for methadone maintenance) and 101 individual control subjects, all Caucasian. Using separate analyses for autosomal and X chromosomal variants, we found that the strongest associations of allele frequency with heroin addiction were with the autosomal variants rs965972, located in the Unigene cluster Hs.147755 (experiment-wise q=0.053), and rs1986513 (q=0.187). The three variants exhibiting the strongest association with heroin addiction by genotype frequency were rs1714984, located in an intron of the gene for the transcription factor myocardin (P=0.000022), rs965972 (P=0.000080) and rs1867898 (P=0.000284). One genotype pattern (AG-TT-GG) was found to be significantly associated with developing heroin addiction (odds ratio (OR)=6.25) and explained 27% of the population attributable risk for heroin addiction in this cohort. Another genotype pattern (GG-CT-GG) of these variants was found to be significantly associated with protection from developing heroin addiction (OR=0.13), and lacking this genotype pattern explained 83% of the population attributable risk for developing heroin addiction. Evidence was found for involvement of five genes in heroin addiction, the genes coding for the mu opioid receptor, the metabotropic receptors mGluR6 and mGluR8, nuclear receptor NR4A2 and cryptochrome 1 (photolyase-like). This approach has identified several new genes potentially associated with heroin addiction and has confirmed the role of OPRM1 in this disease.
Collapse
Affiliation(s)
- D A Nielsen
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Reyes-Gibby CC, Shete S, Rakvåg T, Bhat SV, Skorpen F, Bruera E, Kaasa S, Klepstad P. Exploring joint effects of genes and the clinical efficacy of morphine for cancer pain: OPRM1 and COMT gene. Pain 2007; 130:25-30. [PMID: 17156920 PMCID: PMC1995596 DOI: 10.1016/j.pain.2006.10.023] [Citation(s) in RCA: 229] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Revised: 10/06/2006] [Accepted: 10/24/2006] [Indexed: 11/20/2022]
Abstract
Pain is a complex human trait. It is likely that the interaction of multiple genes, each with a small individual effect, along with the effect of environmental factors, influences the clinical efficacy of opioids rather than a single gene alone. Polymorphisms in genes coding for the mu-opioid receptor (A118G) and catechol-O-methyl transferase (Val158Met) may be important modulators of opioid efficacy. We assessed joint effects of the OPRM1 and COMT genes in predicting morphine dose for cancer pain relief. We used genotype and clinical data from a pharmacokinetic study of morphine in 207 inpatients treated with stable morphine dose for at least 3 days by Palliative Medicine Specialists. Results showed significant variation in morphine dose requirement by genotype groups: carriers of COMT Val/Val and Val/Met genotype required 63% and 23%, respectively, higher morphine dose compared to carriers of Met/Met genotype (p=0.02). Carriers of OPRM1 GG genotype required 93% higher morphine dose compared to carriers of AA genotypes (p=0.012). When we explored for joint effects, we found that carriers of the OPRM1 AA and COMT Met/Met genotype required the lowest morphine dose to achieve pain relief (87 mg/24 h; 95%CI=57,116) and those with neither Met/Met nor AA genotype needed the highest morphine dose (147 mg/24 h; 95%CI=100,180). The significant joint effects for the Met/Met and AA genotypes (p<0.012) persisted, even after controlling for demographic and clinical variables in the multivariable analyses. Future studies are needed to further characterize the joint effects of multiple genes, along with demographic and clinical variables, in predicting opioid dose.
Collapse
|
16
|
Han W, Kasai S, Hata H, Takahashi T, Takamatsu Y, Yamamoto H, Uhl GR, Sora I, Ikeda K. Intracisternal A-particle element in the 3' noncoding region of the mu-opioid receptor gene in CXBK mice: a new genetic mechanism underlying differences in opioid sensitivity. Pharmacogenet Genomics 2006; 16:451-60. [PMID: 16708053 DOI: 10.1097/01.fpc.0000215072.36965.8d] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES CXBK mice, recombinant inbred mice derived from C57BL/6By and BALB/cBy progenitors, display reduced morphine-induced analgesia. Earlier we reported that CXBK mice expressed a reduced amount of the major transcript, MOR-1 mRNA, of the mu-opioid receptor gene. The CXBK MOR-1 mRNA contains a normal coding region and an abnormally long untranslated region. METHODS AND RESULTS To identify the nucleotide-sequence difference between the CXBK MOR-1 mRNA and that of the progenitors, we first characterized the 3' untranslated region of the MOR-1 mRNA, which was largely unknown. A 3' rapid amplification of cDNA ends-PCR analysis revealed that the 3' untranslated region of the C57BL/6By MOR-1 mRNA was 10 181 nucleotides transcribed from an exon. Next, we compared the MOR-1 genes in C57BL/6By, CXBK, and BALB/cBy mice, and found a 5293 nucleotide insertion only in CXBK mice. The inserted sequence was a variant of the intracisternal A-particle elements that exist in the mouse genome at approximately 1000 sites. Reverse transcription-PCR analyses revealed that the intracisternal A-particle element was transcribed as a part of the CXBK MOR-1 mRNA. No other differences were found in the MOR-1 mRNA between CXBK and BALB/cBy mice, whereas 100 nucleotides differed between C57BL/6By and CXBK mice aside from the intracisternal A-particle insertion. Finally, CXBK mice displayed reduced morphine responses compared with BALB/cBy mice. CONCLUSIONS Our data suggest that differences in the MOR-1 3' untranslated region appear to cause the CXBK phenotype. This genetic mechanism underlying the CXBK phenotype may provide good insight into the possible genetic mechanisms underlying individual differences in opioid sensitivity in humans.
Collapse
MESH Headings
- 3' Untranslated Regions
- Analgesics, Opioid/pharmacology
- Animals
- Base Sequence
- Crosses, Genetic
- Drug Resistance/genetics
- Genes, Intracisternal A-Particle
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- Molecular Sequence Data
- Morphine/pharmacology
- RNA, Messenger/chemistry
- Receptors, Opioid, mu/genetics
Collapse
Affiliation(s)
- Wenhua Han
- Division of Psychobiology, Tokyo Institute of Psychiatry, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Pellegrini S, Wood M, Daniel AM, Papini MR. Opioid receptors modulate recovery from consummatory successive negative contrast. Behav Brain Res 2006; 164:239-49. [PMID: 16135386 DOI: 10.1016/j.bbr.2005.06.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Revised: 06/16/2005] [Accepted: 06/16/2005] [Indexed: 01/30/2023]
Abstract
Three experiments explored the role of the opioid system in consummatory successive negative contrast. In Experiment 1, rats treated with the nonspecific opioid-receptor antagonist naloxone (2mg/kg) exhibited increased suppression after a shift from 32% to 6% sucrose solution (32-->6), relative to 6-->6 unshifted controls. A similar but shorter effect was observed with the delta-opioid receptor antagonist naltrindole (1mg/kg). In Experiment 2, naloxone increased suppression after a more conventional 32-->4 sucrose shift. In Experiment 3, rats classified as expressing slow recovery from contrast (after a 32-->4 sucrose downshift) were more sensitive to naloxone in an activity test than fast-recovery rats. Whereas it was previously known that contrast was reduced by the extrinsic administration of opioid agonists, the effects reported here with antagonists provide the first evidence that the opioid system is intrinsically engaged by situations involving surprising reward loss.
Collapse
Affiliation(s)
- Santiago Pellegrini
- Instituto de Biologia y Medicina Experimental and Universidad de Buenos Aires, Argentina
| | | | | | | |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Abstract
OBJECTIVE The completion of the human genome sequence has spurred investigation of the genetic contribution to substance dependence. In this article some of the recent scientific evidence for genetic determinants of opioid and cocaine dependence is reviewed. METHOD An electronic search of the medical literature was conducted to locate published studies relevant to the genetics of opioid and cocaine dependence. The collected information judged to be most pertinent is described and discussed. RESULTS Genetic epidemiologic studies support a high degree of heritable vulnerability for both opioid and cocaine dependence. Polymorphisms in the genes coding for dopamine receptors and transporter, opioid receptors, endogenous opioid peptides, cannabinoid receptors, and serotonin receptors and transporter all appear to be associated with the phenotypic expression of this vulnerability once opioids or cocaine are consumed. CONCLUSIONS Despite this initial progress, identification of specific genes and quantification of associated risk for the expression of each gene remain to be elucidated. While alteration of an individual's genome to change the phenotype seems remote, future interventions for treatment of opioid and cocaine dependence may include precise medications targeted to block the effects of proteins that have been identified through genetic research.
Collapse
Affiliation(s)
- Andrew J Saxon
- Department of Psychiatry, University of Washington School of Medicine; Center of Excellence in Substance Abuse Treatment and Education, VA Puget Sound Health Care System, Seattle, WA 98108, USA.
| | | | | |
Collapse
|
23
|
Zalewska-Kaszubska J, Czarnecka E. Deficit in beta-endorphin peptide and tendency to alcohol abuse. Peptides 2005; 26:701-5. [PMID: 15752586 DOI: 10.1016/j.peptides.2004.11.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2004] [Revised: 11/09/2004] [Accepted: 11/10/2004] [Indexed: 11/24/2022]
Abstract
Human and animal studies suggest that there is a correlation between endogenous opioid peptides, especially beta-endorphin, and alcohol abuse. It has been proven that the consumption of alcohol activates the endogenous opioid system. Consumption of alcohol results in an increase in beta-endorphin level in those regions of the human brain, which are associated with a reward system. However, it has also been observed that habitual alcohol consumption leads to a beta-endorphin deficiency. It is a well-documented phenomenon that people with a genetic deficit of beta-endorphin peptide are particularly susceptible to alcoholism. The plasma level of beta-endorphin in subjects genetically at high risk of excessive alcohol consumption shows lower basal activity of this peptide. Its release increases significantly after alcohol consumption. Clinical and laboratory studies confirm that certain genetically determined factors might increase the individual's vulnerability to alcohol abuse.
Collapse
|
24
|
Kreek MJ, Bart G, Lilly C, LaForge KS, Nielsen DA. Pharmacogenetics and human molecular genetics of opiate and cocaine addictions and their treatments. Pharmacol Rev 2005; 57:1-26. [PMID: 15734726 DOI: 10.1124/pr.57.1.1] [Citation(s) in RCA: 241] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Opiate and cocaine addictions are major social and medical problems that impose a significant burden on society. Despite the size and scope of these problems, there are few effective treatments for these addictions. Methadone maintenance is an effective and most widely used treatment for opiate addiction, allowing normalization of many physiological abnormalities caused by chronic use of short-acting opiates. There are no pharmacological treatments for cocaine addiction. Epidemiological, linkage, and association studies have demonstrated a significant contribution of genetic factors to the addictive diseases. This article reviews the molecular genetics and pharmacogenetics of opiate and cocaine addictions, focusing primarily on genes of the opioid and monoaminergic systems that have been associated with or have evidence for linkage to opiate or cocaine addiction. This evidence has been marshalled either through identification of variant alleles that lead to functional alterations of gene products, altered gene expression, or findings of linkage or association studies. Studies of polymorphisms in the mu opioid receptor gene, which encodes the receptor target of some endogenous opioids, heroin, morphine, and synthetic opioids, have contributed substantially to knowledge of genetic influences on opiate and cocaine addiction. Other genes of the endogenous opioid and monoaminergic systems, particularly genes encoding dopamine beta-hydroxylase, and the dopamine, serotonin, and norepinephrine transporters have also been implicated. Variants in genes encoding proteins involved in metabolism or biotransformation of drugs of abuse and also of treatment agents are reviewed.
Collapse
Affiliation(s)
- Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, Box 171, 1230 York Avenue, New York, New York 10021, USA.
| | | | | | | | | |
Collapse
|
25
|
Ide S, Kobayashi H, Tanaka K, Ujike H, Sekine Y, Ozaki N, Inada T, Harano M, Komiyama T, Yamada M, Iyo M, Ikeda K, Sora I. Gene polymorphisms of the mu opioid receptor in methamphetamine abusers. Ann N Y Acad Sci 2005; 1025:316-24. [PMID: 15542732 DOI: 10.1196/annals.1316.039] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
In drug addiction, the opioid system is thought to mediate motivational effects through dopamine-independent mechanisms. We have investigated associations of the mu-opioid receptor gene (OPRM) variations with methamphetamine (MAP) dependence/psychosis. The allelic frequency of A118G (Asn40Asp) in exon 1 of ORPM was 45.3% in our control subjects, but only 7.5-25.8% in the Caucasian or African-American population of previous studies. We have identified several novel polymorphisms in intron 1 and the 5' untranslated region (5'UTR) of OPRM. Polymorphisms in the functionally relevant 5' regulatory region of OPRM were different in our Japanese population from Caucasian or African-American populations. No significant differences between controls and MAP abusers were found in either genotype or allele frequency at any single nucleotide polymorphism (SNP) or (AC)n dinucleotide repeat in intron 1. A subdivision of our MAP group revealed that A118G of OPRM shows a significant association with MAP psychosis having latency less than three years. Further analysis should be capable of identifying associations between the OPRM variations and MAP dependence/psychosis.
Collapse
Affiliation(s)
- Soichiro Ide
- Department of Molecular Psychiatry, Tokyo Institute of Psychiatry, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Kreek MJ, Schlussman SD, Bart G, Laforge KS, Butelman ER. Evolving perspectives on neurobiological research on the addictions: celebration of the 30th anniversary of NIDA. Neuropharmacology 2004; 47 Suppl 1:324-44. [PMID: 15464148 DOI: 10.1016/j.neuropharm.2004.07.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2004] [Revised: 07/02/2004] [Accepted: 07/20/2004] [Indexed: 01/31/2023]
Abstract
The roots of the Laboratory of the Biology of the Addictive Diseases are in the development of methadone maintenance for the treatment of opiate addiction. Methadone maintenance therapy continues to be one of the major effective forms of addiction pharmacotherapy and underscores the importance of biological factors in the physiology and treatment of the addictive diseases. Recent work in the Laboratory has focused on the neurobiological, neurochemical, neuroendocrine and behavioral aspects of addictive diseases (principally cocaine and the opiate addictions), using an interdisciplinary approach. The models we have focused on range from in vitro molecular biology and neuroscience, to in vivo animal models, to experiments in normal human populations and patients with specific addictive diseases, and most recently to the human molecular genetics of different addictive diseases. Two long-term corollary hypotheses have guided the Laboratory's work: (1) That the endogenous opioid peptide/receptor systems play a central role in the addictive states and therefore in their treatment. (2) That atypical responsivity to stressors (e.g., in the hypothalamic-pituitary-adrenal axis) plays a role in vulnerability and relapse to specific addictive diseases. This atypical responsivity may be drug-induced, environmentally acquired, and/or due to genetic variation.
Collapse
Affiliation(s)
- Mary Jeanne Kreek
- Laboratory of the Biology of the Addictive Diseases, The Rockefeller University, 1230 York Avenue, Box 171, New York, NY 10021, USA.
| | | | | | | | | |
Collapse
|
27
|
Yuferov V, Fussell D, LaForge KS, Nielsen DA, Gordon D, Ho A, Leal SM, Ott J, Kreek MJ. Redefinition of the human kappa opioid receptor gene (OPRK1) structure and association of haplotypes with opiate addiction. PHARMACOGENETICS 2004; 14:793-804. [PMID: 15608558 PMCID: PMC6141019 DOI: 10.1097/00008571-200412000-00002] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The kappa opioid receptor (KOR) plays a role in stress responsivity, opiate withdrawal and responses to cocaine. KOR activation by its endogenous ligand dynorphin A(1-17) decreases basal and drug-induced striatal levels of dopamine. The complete structure of the human KOR gene (hOPRK1) has not been previously determined. This study: (i) characterized the genomic structure of the hOPRK1 gene; (ii) identified single nucleotide polymorphisms (SNPs) in the hOPRK1 gene; and (iii) investigated possible associations of these variants with vulnerability to develop heroin addiction. Analysis of 5'-RACE cDNA clones revealed the presence of a novel exon 1 ranging in length from 167 to 251 nucleotides in the 5' 5'-untranslated region of the hOPRK1 mRNA. We found that the hOPRK1 gene has four major exons and three introns, similar to rodent OPRK1 genes. Direct sequencing of amplified DNA containing all four exons and intron 1 of the hOPRK1 gene were evaluated for polymorphisms in 291 subjects (145 former heroin addicts and 146 controls). Twelve SNPs were identified, nine novel variants and three previously reported SNPs. Using logistic regression with opioid dependence as the dependent variable, the 36G>T SNP exhibited a point-wise significant association (P = 0.016) with disease status. The number of haplotypes seen in the three ethnic groups were nine, six and five for African-Americans, Caucasians, and Hispanics, respectively, with corresponding significance levels for differences in haplotype frequencies between cases and controls of P = 0.0742, 0.1015 and 0.0041. Combining ethnicities by Fisher's method yields an empirical significance level of P = 0.0020.
Collapse
Affiliation(s)
- Vadim Yuferov
- aLaboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10021, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Lee PW, Wu S, Lee YM. Differential Expression of μ-Opioid Receptor Gene in CXBK and B6 Mice by Sp1. Mol Pharmacol 2004. [DOI: 10.1124/mol.66.6.1580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
29
|
|
30
|
Loh EW, Fann CSJ, Chang YT, Chang CJ, Cheng ATA. Endogenous Opioid Receptor Genes and Alcohol Dependence Among Taiwanese Han. Alcohol Clin Exp Res 2004; 28:15-9. [PMID: 14745298 DOI: 10.1097/01.alc.0000106303.41755.b8] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Nonselective opioid antagonists reduce alcohol consumption under various experimental situations, and several association studies have examined possible roles of opioid receptor mu (OPRM), delta (OPRD), and kappa (OPRK) genes in the development of alcohol dependence. METHODS We examined 20 single nucleotide polymorphisms (SNPs) across the OPRM, OPRD, and OPRK genes in 158 alcohol-dependent subjects and 149 controls. Differences in allele frequency and genotype distribution between case subjects and controls, as well as the deviation from Hardy-Weinberg equilibrium, were examined using Fisher's exact tests. RESULTS No significant difference in either allele or genotype frequency was found between case subjects and controls for each of the SNPs. CONCLUSIONS Our findings do not support a possible role of the opioid receptor genes for the proclivity to alcohol dependence in the Taiwanese Han.
Collapse
Affiliation(s)
- El Wui Loh
- National Genotyping Center at Academia Sinica, Academia Sinica, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
31
|
Compton P, Geschwind DH, Alarcón M. Association between human mu-opioid receptor gene polymorphism, pain tolerance, and opioid addiction. Am J Med Genet B Neuropsychiatr Genet 2003; 121B:76-82. [PMID: 12898579 DOI: 10.1002/ajmg.b.20057] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Central to both pain responses and opioid addiction is activity at the micro -opioid receptor. To explore the role of the micro -opioid receptor gene (OPRM) in human pain tolerance and opioid addiction, we examined the relationships among OPRM genotype and experimental pain tolerance in opioid addicts in methadone treatment (n = 50) and healthy normal controls (n = 59). Pain phenotype (pain tolerant vs. pain intolerant) was operationalized as tolerance to a standardized noxious stimulus (either thermal or mechanical), and dichotomized based on distribution. One microsatellite and two single nucleotide polymorphisms, A118G and C17T, in exon 1 were typed to study the OPRM gene. Although the established relationship between the phenotypes of opioid addiction and pain intolerance was validated (P = 0.02), genotype differed neither between addict-affected vs. control, nor pain tolerant vs. intolerant subjects. The variant A118G was absent in all individuals and the C17T polymorphism appeared in only three African-American individuals (two addicts and one control). The absence of this polymorphism, the small sample size and the heterogeneous ethnic backgrounds of participants in the pilot study allow only tentative conclusions based on the results, thus the role of the opioid receptor in pain and opioid reward response remains uncertain.
Collapse
Affiliation(s)
- Peggy Compton
- Acute Care Section, UCLA School of Nursing, Los Angeles, California 90095-6918, USA.
| | | | | |
Collapse
|
32
|
Franke P, Wendel B, Knapp M, Schwab SG, Neef D, Maier W, Wildenauer DB, Hoehe MR. Introducing a new recruitment approach to sample collection for genetic association studies in opioid dependence. Eur Psychiatry 2003; 18:18-22. [PMID: 12648891 DOI: 10.1016/s0924-9338(02)00005-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVE In a modified case-control association study we tested the assumption that two polymorphisms (A(118)G in exon 1 and IVS2+31 in intron 2) of the human mu-opioid receptor gene (OPRM1) confer susceptibility to opioid dependence. METHODS In contrast to classical case-control studies both groups, opioid dependent cases and non-opioid dependent controls were recruited from individuals who have had access to drugs including opioids and who had been sentenced for violation of the "Dangerous Drugs Act" in Germany. RESULTS For the two allelic variants of OPRM1 under study we did not find evidence for association with opioid dependence. CONCLUSIONS ;Despite absence of association we think that this recruitment approach introduced here, is useful since it putatively offers a more adequate matching for case-control association studies of opioid dependent individuals.
Collapse
Affiliation(s)
- Petra Franke
- Department of Psychiatry, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Recent discoveries in opioid pharmacology help explain the enormous variability in clinical responses to these powerful analgesics. Although there is only one m opioid receptor gene, splice variants of that gene's expression result in a panoply of different functioning receptors. Other sources of variable response include polymorphisms in the m opioid receptor regulatory region, and pharmacokinetic differences because of cytochrome P-450 mono-oxygenase heterogeneity. Analgesic tolerance is likely the key phenomenon limiting the benefit of opioids. A plethora of intracellular pathways affects this. Among them are the N-methyl-D-aspartate receptor, protein kinase C gamma activity, nitric oxide synthase, and GM1 ganglioside content of the neuronal membrane. Clinical studies undercut the routine use of meperidine in most settings. Other studies have shown better ways to diminish opioid side effects.
Collapse
MESH Headings
- Analgesics, Opioid/adverse effects
- Analgesics, Opioid/therapeutic use
- Animals
- Clinical Trials as Topic
- Drug Tolerance
- Humans
- Pain/drug therapy
- Pain/physiopathology
- Receptors, Opioid/drug effects
- Receptors, Opioid/genetics
- Receptors, Opioid/physiology
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/physiology
- Trans-Activators/genetics
Collapse
Affiliation(s)
- Eric Chevlen
- Cancer Care Center, St. Elizabeth Hospital, 1044 Belmont Avenue, Youngstown, OH 44501, USA.
| |
Collapse
|
34
|
Börner C, Höllt V, Kraus J. Involvement of activator protein-1 in transcriptional regulation of the human mu-opioid receptor gene. Mol Pharmacol 2002; 61:800-5. [PMID: 11901219 DOI: 10.1124/mol.61.4.800] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
mu-Opioid receptors mediate such opioid effects as analgesia, euphoria, and immunomodulation. Gene expression of mu-opioid receptors can be modulated by various substances, including cytokines, hormones, and drugs. Some of these stimuli (e.g., IL-1beta and cocaine) have been shown to activate members of the AP-1 transcription factor family. In addition, transcription of the mu-opioid receptor gene is induced by the phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA), an activator of protein kinase C, which in turn is an activator of AP-1 transcription factors. This indicates that signaling pathways involving protein kinase C and activator protein 1 (AP-1) transcription factors are important for the specific expression pattern of the mu-opioid receptor gene. In this report, we show that TPA activates AP-1 as well as the transcription factor nuclear factor kappaB (NFkappaB) in the mu-opioid receptor expressing neuroblastoma cell line SH SY5Y. In transfection experiments performed in these cells, both factors trans-activate expression of reporter gene constructs containing the human mu-opioid receptor gene promoter. By excluding the effects of TPA on NFkappaB with the specific NFkappaB inhibitor sulfasalazine, AP-1 regulatory elements were localized. Two AP-1 elements, which differ in one nucleotide each from the classic AP-1 binding site, were delineated to positions -2388 and -1434 of the promoter. Independent of their orientation, these elements conferred TPA responsiveness on the heterologous thymidine kinase promoter. AP-1 binding to these elements was confirmed using electrophoretic mobility shift and immunoshift assays.
Collapse
Affiliation(s)
- Christine Börner
- Department of Pharmacology, University of Magdeburg, Magdeburg, Germany
| | | | | |
Collapse
|