1
|
Giratallah H, Chenoweth MJ, Pouget JG, El-Boraie A, Alsaafin A, Lerman C, Knight J, Tyndale RF. CYP2A6 associates with respiratory disease risk and younger age of diagnosis: a phenome-wide association Mendelian Randomization study. Hum Mol Genet 2024; 33:198-210. [PMID: 37802914 PMCID: PMC10772040 DOI: 10.1093/hmg/ddad172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 09/21/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023] Open
Abstract
CYP2A6, a genetically variable enzyme, inactivates nicotine, activates carcinogens, and metabolizes many pharmaceuticals. Variation in CYP2A6 influences smoking behaviors and tobacco-related disease risk. This phenome-wide association study examined associations between a reconstructed version of our weighted genetic risk score (wGRS) for CYP2A6 activity with diseases in the UK Biobank (N = 395 887). Causal effects of phenotypic CYP2A6 activity (measured as the nicotine metabolite ratio: 3'-hydroxycotinine/cotinine) on the phenome-wide significant (PWS) signals were then estimated in two-sample Mendelian Randomization using the wGRS as the instrument. Time-to-diagnosis age was compared between faster versus slower CYP2A6 metabolizers for the PWS signals in survival analyses. In the total sample, six PWS signals were identified: two lung cancers and four obstructive respiratory diseases PheCodes, where faster CYP2A6 activity was associated with greater disease risk (Ps < 1 × 10-6). A significant CYP2A6-by-smoking status interaction was found (Psinteraction < 0.05); in current smokers, the same six PWS signals were found as identified in the total group, whereas no PWS signals were found in former or never smokers. In the total sample and current smokers, CYP2A6 activity causal estimates on the six PWS signals were significant in Mendelian Randomization (Ps < 5 × 10-5). Additionally, faster CYP2A6 metabolizer status was associated with younger age of disease diagnosis for the six PWS signals (Ps < 5 × 10-4, in current smokers). These findings support a role for faster CYP2A6 activity as a causal risk factor for lung cancers and obstructive respiratory diseases among current smokers, and a younger onset of these diseases. This research utilized the UK Biobank Resource.
Collapse
Affiliation(s)
- Haidy Giratallah
- Department of Pharmacology and Toxicology, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Campbell Family Mental Health Research Institute, CAMH, 250 College St, Toronto, ON M5T 1R8, Canada
| | - Meghan J Chenoweth
- Department of Pharmacology and Toxicology, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Campbell Family Mental Health Research Institute, CAMH, 250 College St, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Jennie G Pouget
- Campbell Family Mental Health Research Institute, CAMH, 250 College St, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Ahmed El-Boraie
- Department of Pharmacology and Toxicology, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Campbell Family Mental Health Research Institute, CAMH, 250 College St, Toronto, ON M5T 1R8, Canada
| | - Alaa Alsaafin
- Department of Pharmacology and Toxicology, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Campbell Family Mental Health Research Institute, CAMH, 250 College St, Toronto, ON M5T 1R8, Canada
| | - Caryn Lerman
- Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Ave, Los Angeles, CA 90033, United States
| | - Jo Knight
- Department of Psychiatry, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Data Science Institute, Lancaster University Medical School, Lancaster LA1 4YE, United Kingdom
| | - Rachel F Tyndale
- Department of Pharmacology and Toxicology, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Campbell Family Mental Health Research Institute, CAMH, 250 College St, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
2
|
Nasrin S, Coates S, Bardhi K, Watson C, Muscat JE, Lazarus P. Inhibition of Nicotine Metabolism by Cannabidiol (CBD) and 7-Hydroxycannabidiol (7-OH-CBD). Chem Res Toxicol 2023; 36:177-187. [PMID: 36626330 PMCID: PMC9945182 DOI: 10.1021/acs.chemrestox.2c00259] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Indexed: 01/11/2023]
Abstract
Cannabis-based products have experienced notable increases in co-usage alongside tobacco products. Several cannabinoids exhibit inhibition of a number of cytochrome P450 (CYP) and UDP glucuronosyltransferase (UGT) enzymes, but few studies have examined their inhibition of enzymes involved in nicotine metabolism. The goal of the present study was to examine potential drug-drug interactions occurring in the nicotine metabolism pathway perpetrated by cannabidiol (CBD) and its active metabolite, 7-hydroxy-CBD (7-OH-CBD). The inhibitory effects of CBD and 7-OH-CBD were tested in microsomes from HEK293 cells overexpressing individual metabolizing enzymes and from human liver tissue. Assays with overexpressing microsomes demonstrated that CBD and 7-OH-CBD inhibited CYP-mediated nicotine metabolism. Binding-corrected IC50,u values for CBD inhibition of nicotine metabolism to cotinine and nornicotine, and cotinine metabolism to trans-3'-hydroxycotinine (3HC), were 0.27 ± 0.060, 0.23 ± 0.14, and 0.21 ± 0.14 μM, respectively, for CYP2A6; and 0.26 ± 0.17 and 0.029 ± 0.0050 μM for cotinine and nornicotine formation, respectively, for CYP2B6. 7-OH-CBD IC50,u values were 0.45 ± 0.18, 0.16 ± 0.08, and 0.78 ± 0.23 μM for cotinine, nornicotine, and 3HC formation, respectively, for CYP2A6, and 1.2 ± 0.44 and 0.11 ± 0.030 μM for cotinine and nornicotine formation, respectively, for CYP2B6. Similar IC50,u values were observed in HLM. Inhibition (IC50,u = 0.37 ± 0.06 μM) of 3HC to 3HC-glucuronide formation by UGT1A9 was demonstrated by CBD. Significant inhibition of nicotine metabolism pathways by CBD and 7-OH-CBD suggests that cannabinoids may inhibit nicotine metabolism, potentially impacting tobacco addiction and cessation.
Collapse
Affiliation(s)
- Shamema Nasrin
- Department
of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical
Sciences, Washington State University, Spokane, Washington99223, United States
| | - Shelby Coates
- Department
of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical
Sciences, Washington State University, Spokane, Washington99223, United States
| | - Keti Bardhi
- Department
of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical
Sciences, Washington State University, Spokane, Washington99223, United States
| | - Christy Watson
- Department
of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical
Sciences, Washington State University, Spokane, Washington99223, United States
| | - Joshua E. Muscat
- Penn
State Cancer Institute, Department of Public Health Sciences, Penn State University College of Medicine, Hershey, Pennsylvania17033, United States
| | - Philip Lazarus
- Department
of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical
Sciences, Washington State University, Spokane, Washington99223, United States
| |
Collapse
|
3
|
Jones SK, Wolf BJ, Froeliger B, Wallace K, Carpenter MJ, Alberg AJ. Nicotine metabolism predicted by CYP2A6 genotypes in relation to smoking cessation: A systematic review. Nicotine Tob Res 2021; 24:633-642. [PMID: 34478556 DOI: 10.1093/ntr/ntab175] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 09/02/2021] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Identifying genetic factors associated with smoking cessation could inform precision cessation interventions. Of major interest is genetic variation in nicotine metabolism, largely predicted by CYP2A6 variations. METHODS We conducted a systematic literature review to summarize the population-based evidence of the association between CYP2A6 and smoking cessation.In the 12 studies meeting the inclusion criteria, the known functional metabolic effect of CYP2A6 variants was used to classify nicotine metabolism as normal (>75% metabolic activity), intermediate (50.1 - 75% activity), slow (25 - 50% activity), and poor (<25% activity). Summary odds ratios of smoking cessation were calculated across metabolic groups, stratified by ancestry and whether participants received pharmacotherapy or placebo/no treatment. RESULTS Among untreated people of European ancestry (n = 4 studies), those with CYP2A6 reduced metabolism were more likely to quit smoking than those with normal metabolism [Summary OR = 2.05, 95% CI 1.23 - 3.42] and the likelihood of cessation increased as nicotine metabolism decreased. Nicotine replacement therapy attenuated the association at end-of-treatment, while bupropion modified the association such that intermediate/slow metabolizers were less likely to quit than normal metabolizers [Summary OR = 0.86, 95% CI 0.79 - 0.94]. Among untreated Asian people (n = 3 studies), results differed compared to those with European ancestry: those with slow metabolism were less likely to have quit smoking than normal metabolizers [Summary OR = 0.52, 95% CI 0.38 - 0.71]. Evidence for people of African ancestry (n = 1 study) suggested the CYP2A6 association with cessation may differ compared to those of European ancestry. IMPLICATIONS Most studies included in this review were of European ancestry populations; these showed slower nicotine metabolism was associated with increased likelihood of smoking cessation in a dose-related manner. Pharmacotherapy appeared to attenuate or modify this association among people of European ancestry, but it is unclear whether the change in the association remains consistent after treatment ceases. This finding has implications for precision medicine cessation interventions. Based on only a few studies of people of Asian or African ancestry, the association between CYP2A6 variants and cessation may differ from that observed among those of European ancestry, but more evidence is needed.
Collapse
Affiliation(s)
- Stephanie K Jones
- Department of Public Health Sciences, Medical University of South Carolina, Charleston SC
| | - Bethany J Wolf
- Department of Public Health Sciences, Medical University of South Carolina, Charleston SC
| | - Brett Froeliger
- Department of Psychological Sciences, University of Missouri, Columbia MO
| | - Kristin Wallace
- Department of Public Health Sciences, Medical University of South Carolina, Charleston SC.,Hollings Cancer Center, 86 Jonathan Lucas Street, Medical University of South Carolina, Charleston SC
| | - Matthew J Carpenter
- Hollings Cancer Center, 86 Jonathan Lucas Street, Medical University of South Carolina, Charleston SC.,Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston SC
| | - Anthony J Alberg
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia SC
| |
Collapse
|
4
|
Murphy SE. Biochemistry of nicotine metabolism and its relevance to lung cancer. J Biol Chem 2021; 296:100722. [PMID: 33932402 PMCID: PMC8167289 DOI: 10.1016/j.jbc.2021.100722] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/27/2022] Open
Abstract
Nicotine is the key addictive constituent of tobacco. It is not a carcinogen, but it drives smoking and the continued exposure to the many carcinogens present in tobacco. The investigation into nicotine biotransformation has been ongoing for more than 60 years. The dominant pathway of nicotine metabolism in humans is the formation of cotinine, which occurs in two steps. The first step is cytochrome P450 (P450, CYP) 2A6–catalyzed 5′-oxidation to an iminium ion, and the second step is oxidation of the iminium ion to cotinine. The half-life of nicotine is longer in individuals with low P450 2A6 activity, and smokers with low activity often decrease either the intensity of their smoking or the number of cigarettes they use compared with those with “normal” activity. The effect of P450 2A6 activity on smoking may influence one's tobacco-related disease risk. This review provides an overview of nicotine metabolism and a summary of the use of nicotine metabolite biomarkers to define smoking dose. Some more recent findings, for example, the identification of uridine 5′-diphosphoglucuronosyltransferase 2B10 as the catalyst of nicotine N-glucuronidation, are discussed. We also describe epidemiology studies that establish the contribution of nicotine metabolism and CYP2A6 genotype to lung cancer risk, particularly with respect to specific racial/ethnic groups, such as those with Japanese, African, or European ancestry. We conclude that a model of nicotine metabolism and smoking dose could be combined with other lung cancer risk variables to more accurately identify former smokers at the highest risk of lung cancer and to intervene accordingly.
Collapse
Affiliation(s)
- Sharon E Murphy
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
5
|
El‐Boraie A, Taghavi T, Chenoweth MJ, Fukunaga K, Mushiroda T, Kubo M, Lerman C, Nollen NL, Benowitz NL, Tyndale RF. Evaluation of a weighted genetic risk score for the prediction of biomarkers of CYP2A6 activity. Addict Biol 2020; 25:e12741. [PMID: 30815984 DOI: 10.1111/adb.12741] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 12/01/2018] [Accepted: 12/16/2018] [Indexed: 12/12/2022]
Abstract
The nicotine metabolite ratio (NMR; 3-hydroxycotinine/cotinine) is an index of CYP2A6 activity. CYP2A6 is responsible for nicotine's metabolic inactivation and variation in the NMR/CYP2A6 is associated with several smoking behaviors. Our aim was to integrate established alleles and novel genome-wide association studies (GWAS) signals to create a weighted genetic risk score (wGRS) for the CYP2A6 gene for European-ancestry populations. The wGRS was compared with a previous CYP2A6 gene scoring approach designed for an alternative phenotype (C2/N2; cotinine-d2/(nicotine-d2 + cotinine-d2)). CYP2A6 genotypes and the NMR were assessed in European-ancestry participants. The wGRS training set included N = 933 smokers recruited to the Pharmacogenetics of Nicotine Addiction and Treatment clinical trial [NCT01314001]. The replication cohort included N = 196 smokers recruited to the Quit 2 Live clinical trial [NCT01836276]. Comparisons between the two CYP2A6 phenotypes and with fractional clearance were made in a laboratory-based pharmacokinetic study (N = 92 participants). In both the training and replication sets, the wGRS, which included seven CYP2A6 variants, explained 33.8% (P < 0.001) of the variance in NMR, providing improved predictive power to the NMR phenotype when compared with other CYP2A6 gene scoring approaches. NMR and C2/N2 were strongly correlated to nicotine clearance (ρ = 0.70 and ρ = 0.79, respectively; P < 0.001), and to one another (ρ = 0.82; P < 0.001); however reduced function genotypes occurred in slow NMR but throughout C2/N2. The wGRS was able to predict smoking quantity and nicotine intake, to discriminate between NMR slow and normal metabolizers (AUC = 0.79; P < 0.001), and to replicate previous NMR-stratified cessation outcomes showing unique treatment outcomes between metabolizer groups.
Collapse
Affiliation(s)
- Ahmed El‐Boraie
- Department of Pharmacology and ToxicologyUniversity of Toronto Toronto M5S 1A8 Canada
| | - Taraneh Taghavi
- Department of Pharmacology and ToxicologyUniversity of Toronto Toronto M5S 1A8 Canada
| | - Meghan J. Chenoweth
- Department of Pharmacology and ToxicologyUniversity of Toronto Toronto M5S 1A8 Canada
| | - Koya Fukunaga
- Center for Integrative Medical SciencesRIKEN Yokohama Kanagawa 230‐0045 Japan
| | - Taisei Mushiroda
- Center for Integrative Medical SciencesRIKEN Yokohama Kanagawa 230‐0045 Japan
| | - Michiaki Kubo
- Center for Integrative Medical SciencesRIKEN Yokohama Kanagawa 230‐0045 Japan
| | - Caryn Lerman
- Department of Psychiatry and Abramson Cancer CenterUniversity of Pennsylvania Philadelphia 19104 Pennsylvania
| | - Nicole L. Nollen
- Department of Preventive Medicine and Public HealthUniversity of Kansas Kansas City 66160 Kansas
| | - Neal L. Benowitz
- Departments of Medicine and Biopharmaceutical Sciences, Division of Clinical Pharmacology and Experimental Therapeutics, Medical Services and Center for Tobacco Control Research and EducationUniversity of California San Francisco 94110 California
| | - Rachel F. Tyndale
- Department of Pharmacology and ToxicologyUniversity of Toronto Toronto M5S 1A8 Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health and Division of Brain and Therapeutics, Department of PsychiatryUniversity of Toronto Toronto M6J 1H4 Canada
| |
Collapse
|
6
|
Prevalence of Some Genetic Risk Factors for Nicotine Dependence in Ukraine. GENETICS RESEARCH INTERNATIONAL 2019; 2019:2483270. [PMID: 31885928 PMCID: PMC6925678 DOI: 10.1155/2019/2483270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/22/2019] [Indexed: 11/17/2022]
Abstract
Tobacco smoking is known to be a strong risk factor for developing many diseases. The development and severity of smoking dependence results from interaction of environmental and lifestyle factors, psycho-emotional predispositions, and also from genetic susceptibility. In present study, we investigated polymorphic variants in genes contributed to nicotine dependence, as well as to increased impulsivity, known to be an important risk factor for substance use disorders, in Ukraine population. The genotype frequencies at CYP2A6, DNMT3B, DRD2, HTR2A, COMT, BDNF, GABRA2, CHRNA5, and DAT1 polymorphisms were determined in 171 Ukraine residents, and these data were compared with data for several other European populations and main ethnic groups. It has been found that genotype frequencies for all studied loci are in Hardy-Weinberg equilibrium in the Ukrainian population and correspond to the respective frequencies in European populations. These findings suggest a similar impact of these loci on nicotine dependence in Ukraine. Further studies with larger sample sizes are, however, needed to draw firm conclusions about the effect size of these polymorphisms.
Collapse
|
7
|
Bloom AJ, Upadhyaya P, Kharasch ED. Strain-specific altered nicotine metabolism in 3,3'-diindolylmethane (DIM) exposed mice. Biopharm Drug Dispos 2019; 40:188-194. [PMID: 31016737 DOI: 10.1002/bdd.2182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/02/2019] [Accepted: 04/08/2019] [Indexed: 12/25/2022]
Abstract
Two indole compounds, indole-3-carbinol (I3C) and its acid condensation product, 3,3'-diindolymethane (DIM), have been shown to suppress the expression of flavin-containing monooxygenases (FMO) and to induce some hepatic cytochrome P450s (CYPs) in rats. In liver microsomes prepared from rats fed I3C or DIM, FMO-mediated nicotine N-oxygenation was decreased, whereas CYP-mediated nicotine metabolism to nicotine iminium and subsequently to cotinine was unchanged. Therefore, it was hypothesized that in mice DIM would also suppress nicotine N-oxygenation without affecting CYP-mediated nicotine metabolism. Liver microsomes were produced from male and female C57BL/6 J and CD1 mice fed 2500 parts per million (ppm) DIM for 14 days. In liver microsomes from DIM-fed mice, FMO-mediated nicotine N-oxygenation did not differ from the controls, but CYP-mediated nicotine metabolism was significantly increased, with results varying by sex and strain. To confirm the effects of DIM in vivo, control and DIM-fed CD1 male mice were injected subcutaneously with nicotine, and the plasma concentrations of nicotine, cotinine and nicotine-N-oxide were measured over 30 minutes. The DIM-fed mice showed greater cotinine concentrations compared with the controls 10 minutes following injection. It is concluded that the effects of DIM on nicotine metabolism in vitro and in vivo differ between mice and rats and between mouse strains, and that DIM is an effective inducer of CYP-mediated nicotine metabolism in commonly studied mouse strains.
Collapse
Affiliation(s)
- A Joseph Bloom
- Department of Genetics, Washington University, St Louis, MO
| | | | - Evan D Kharasch
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC
| |
Collapse
|
8
|
Bloom AJ, Wang P, Kharasch ED. Nicotine oxidation by genetic variants of CYP2B6 and in human brain microsomes. Pharmacol Res Perspect 2019; 7:e00468. [PMID: 30906561 PMCID: PMC6411694 DOI: 10.1002/prp2.468] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 01/20/2019] [Indexed: 11/16/2022] Open
Abstract
Common variation in the CYP2B6 gene, encoding the cytochrome P450 2B6 enzyme, is associated with substrate-specific altered clearance of multiple drugs. CYP2B6 is a minor contributor to hepatic nicotine metabolism, but the enzyme has been proposed as relevant to nicotine-related behaviors because of reported CYP2B6 mRNA expression in human brain tissue. Therefore, we hypothesized that CYP2B6 variants would be associated with altered nicotine oxidation, and that nicotine metabolism by CYP2B6 would be detected in human brain microsomes. We generated recombinant enzymes in insect cells corresponding to nine common CYP2B6 haplotypes and demonstrate genetically determined differences in nicotine oxidation to nicotine iminium ion and nornicotine for both (S) and (R)-nicotine. Notably, the CYP2B6.6 and CYP2B6.9 variants demonstrated lower intrinsic clearance relative to the reference enzyme, CYP2B6.1. In the presence of human brain microsomes, along with nicotine-N-oxidation, we also detect nicotine oxidation to nicotine iminium ion. However, unlike N-oxidation, this activity is NADPH independent, does not follow Michaelis-Menten kinetics, and is not inhibited by NADP or carbon monoxide. Furthermore, metabolism of common CYP2B6 probe substrates, methadone and ketamine, is not detected in the presence of brain microsomes. We conclude that CYP2B6 metabolizes nicotine stereoselectively and common CYP2B6 variants differ in nicotine metabolism activity, but did not find evidence of CYP2B6 activity in human brain.
Collapse
Affiliation(s)
- Adam Joseph Bloom
- Department of Psychiatry and AnesthesiologyWashington UniversitySt. LouisMissouri
| | - Pan‐Fen Wang
- Department of AnesthesiologyDuke University School of MedicineDurhamNorth Carolina
| | - Evan D. Kharasch
- Department of AnesthesiologyDuke University School of MedicineDurhamNorth Carolina
| |
Collapse
|
9
|
Chen LS, Hartz SM, Baker TB, Ma Y, L Saccone N, Bierut LJ. Use of polygenic risk scores of nicotine metabolism in predicting smoking behaviors. Pharmacogenomics 2018; 19:1383-1394. [PMID: 30442082 PMCID: PMC6562697 DOI: 10.2217/pgs-2018-0081] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/10/2018] [Indexed: 01/19/2023] Open
Abstract
AIM This study tests whether polygenic risk scores (PRSs) for nicotine metabolism predict smoking behaviors in independent data. MATERIALS & METHODS Linear regression, logistic regression and survival analyses were used to analyze nicotine metabolism PRSs and nicotine metabolism, smoking quantity and smoking cessation. RESULTS Nicotine metabolism PRSs based on two genome wide association studies (GWAS) meta-analyses significantly predicted nicotine metabolism biomarkers (R2 range: 9.2-16%; minimum p = 7.6 × 10-8). The GWAS top hit variant rs56113850 significantly predicted nicotine metabolism biomarkers (R2 range: 14-17%; minimum p = 4.4 × 10-8). There was insufficient evidence for these PRSs predicting smoking quantity and smoking cessation. CONCLUSION Results suggest that nicotine metabolism PRSs based on GWAS meta-analyses predict an individual's nicotine metabolism, so does use of the top hit variant. We anticipate that PRSs will enter clinical medicine, but additional research is needed to develop a more comprehensive genetic score to predict smoking behaviors.
Collapse
Affiliation(s)
- Li-Shiun Chen
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO 63110, USA
| | - Sarah M Hartz
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Timothy B Baker
- Department of Medicine, University of Wisconsin School of Medicine & Public Health, Madison, WI 53711, USA
| | - Yinjiao Ma
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Nancy L Saccone
- Department of Genetics, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Laura J Bierut
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO 63110, USA
| |
Collapse
|
10
|
Salloum NC, Buchalter ELF, Chanani S, Espejo G, Ismail MS, Laine RO, Nageeb M, Srivastava AB, Trapp N, Trillo L, Vance E, Wenzinger M, Hartz SM, David SP, Chen LS. From genes to treatments: a systematic review of the pharmacogenetics in smoking cessation. Pharmacogenomics 2018; 19:861-871. [PMID: 29914292 PMCID: PMC6219447 DOI: 10.2217/pgs-2018-0023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 04/30/2018] [Indexed: 12/12/2022] Open
Abstract
Smoking cessation treatment outcomes may be heavily influenced by genetic variations among smokers. Therefore, identifying specific variants that affect response to different pharmacotherapies is of major interest to the field. In the current study, we systematically review all studies published in or after the year 1990 which examined one or more gene-drug interactions for smoking cessation treatment. Out of 644 citations, 46 articles met the inclusion criteria for the systematic review. We summarize evidence on several genetic polymorphisms (CHRNA5-A3-B4, CYP2A6, DBH, CHRNA4, COMT, DRD2, DRD4 and CYP2B6) and their potential moderating pharamacotherarpy effects on patient cessation efficacy rates. These findings are promising and call for further research to demonstrate the effectiveness of genetic testing in personalizing treatment decision-making and improving outcome.
Collapse
Affiliation(s)
- Naji C Salloum
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Erica LF Buchalter
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Swati Chanani
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Gemma Espejo
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Mahjabeen S Ismail
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Randy O Laine
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Maysaa Nageeb
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - A Benjamin Srivastava
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Nicholas Trapp
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Ludwig Trillo
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Erica Vance
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Michael Wenzinger
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Sarah M Hartz
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Sean P David
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Li-Shiun Chen
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
11
|
Chen LS, Horton A, Bierut L. Pathways to precision medicine in smoking cessation treatments. Neurosci Lett 2018; 669:83-92. [PMID: 27208830 PMCID: PMC5115988 DOI: 10.1016/j.neulet.2016.05.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/12/2016] [Accepted: 05/17/2016] [Indexed: 02/06/2023]
Abstract
Cigarette smoking is highly addictive and modern genetic research has identified robust genetic influences on nicotine dependence. An important step in translating these genetic findings to clinical practice is identifying the genetic factors affecting smoking cessation in order to enhance current smoking cessation treatments. We reviewed the significant genetic variants that predict nicotine dependence, smoking cessation, and response to cessation pharmacotherapy. These data suggest that genetic risks can predict smoking cessation outcomes and moderate the effect of pharmacological treatments. Some pharmacogenetic findings have been replicated in meta-analyses or in multiple smoking cessation trials. The variation in efficacy between smokers with different genetic markers supports the notion that personalized smoking cessation intervention based upon genotype could maximize the efficiency of such treatment while minimizing side effects, thus influencing the number needed to treat (NNT) and the number needed to harm. In summary, as precision medicine is revolutionizing healthcare, smoking cessation may be one of the first areas where genetic variants may identify individuals at increased risk. Current evidence strongly suggests that genetic variants predict cessation failure and that cessation pharmacotherapy effectiveness is modulated by biomarkers such as nicotinic cholinergic receptor α5 subunit (CHRNA5) genotypes or nicotine metabolism ratio (NMR). These findings strengthen the case for the development and rigorous testing of treatments that target patients with different biological risk profiles.
Collapse
Affiliation(s)
- Li-Shiun Chen
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, United States.
| | - Amy Horton
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Laura Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, United States
| |
Collapse
|
12
|
Saccone NL, Baurley JW, Bergen AW, David SP, Elliott HR, Foreman MG, Kaprio J, Piasecki TM, Relton CL, Zawertailo L, Bierut LJ, Tyndale RF, Chen LS, Genetics and Treatment Networks of the Society for Research on Nicotine and Tobacco (SRNT). The Value of Biosamples in Smoking Cessation Trials: A Review of Genetic, Metabolomic, and Epigenetic Findings. Nicotine Tob Res 2018; 20:403-413. [PMID: 28472521 PMCID: PMC5896536 DOI: 10.1093/ntr/ntx096] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 05/01/2017] [Indexed: 02/03/2023]
Abstract
Introduction Human genetic research has succeeded in definitively identifying multiple genetic variants associated with risk for nicotine dependence and heavy smoking. To build on these advances, and to aid in reducing the prevalence of smoking and its consequent health harms, the next frontier is to identify genetic predictors of successful smoking cessation and also of the efficacy of smoking cessation treatments ("pharmacogenomics"). More broadly, additional biomarkers that can be quantified from biosamples also promise to aid "Precision Medicine" and the personalization of treatment, both pharmacological and behavioral. Aims and Methods To motivate ongoing and future efforts, here we review several compelling genetic and biomarker findings related to smoking cessation and treatment. Results These Key results involve genetic variants in the nicotinic receptor subunit gene CHRNA5, variants in the nicotine metabolism gene CYP2A6, and the nicotine metabolite ratio. We also summarize reports of epigenetic changes related to smoking behavior. Conclusions The results to date demonstrate the value and utility of data generated from biosamples in clinical treatment trial settings. This article cross-references a companion paper in this issue that provides practical guidance on how to incorporate biosample collection into a planned clinical trial and discusses avenues for harmonizing data and fostering consortium-based, collaborative research on the pharmacogenomics of smoking cessation. Implications Evidence is emerging that certain genotypes and biomarkers are associated with smoking cessation success and efficacy of smoking cessation treatments. We review key findings that open potential avenues for personalizing smoking cessation treatment according to an individual's genetic or metabolic profile. These results provide important incentive for smoking cessation researchers to collect biosamples and perform genotyping in research studies and clinical trials.
Collapse
Affiliation(s)
- Nancy L Saccone
- Department of Genetics and Division of Biostatistics, Washington University School of Medicine, St. Louis, MO
| | | | | | - Sean P David
- Department of Medicine, Stanford University, Stanford, CA
| | - Hannah R Elliott
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Marilyn G Foreman
- Pulmonary and Critical Care Medicine, Morehouse School of Medicine, Atlanta, GA
| | - Jaakko Kaprio
- Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - Thomas M Piasecki
- Department of Psychological Sciences, University of Missouri, Columbia, MO
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Laurie Zawertailo
- Nicotine Dependence Service, Centre for Addiction and Mental Health, and Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Laura J Bierut
- Siteman Cancer Center, Institute of Public Health, and Department of Psychiatry, Washington University School of Medicine, St. Louis, MO
| | - Rachel F Tyndale
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, and Departments of Pharmacology & Toxicology and Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Li-Shiun Chen
- Siteman Cancer Center, Institute of Public Health, and Department of Psychiatry, Washington University School of Medicine, St. Louis, MO
| | | |
Collapse
|
13
|
Teitelbaum AM, Murphy SE, Akk G, Baker TB, Germann A, von Weymarn LB, Bierut LJ, Goate A, Kharasch ED, Bloom AJ. Nicotine dependence is associated with functional variation in FMO3, an enzyme that metabolizes nicotine in the brain. THE PHARMACOGENOMICS JOURNAL 2018; 18:136-143. [PMID: 28290528 PMCID: PMC5599305 DOI: 10.1038/tpj.2016.92] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/20/2016] [Accepted: 12/08/2016] [Indexed: 12/16/2022]
Abstract
A common haplotype of the flavin-containing monooxygenase gene FMO3 is associated with aberrant mRNA splicing, a twofold reduction in in vivo nicotine N-oxidation and reduced nicotine dependence. Tobacco remains the largest cause of preventable mortality worldwide. CYP2A6, the primary hepatic nicotine metabolism gene, is robustly associated with cigarette consumption but other enzymes contribute to nicotine metabolism. We determined the effects of common variants in FMO3 on plasma levels of nicotine-N-oxide in 170 European Americans administered deuterated nicotine. The polymorphism rs2266780 (E308G) was associated with N-oxidation of both orally administered and ad libitum smoked nicotine (P⩽3.3 × 10-5 controlling for CYP2A6 genotype). In vitro, the FMO3 G308 variant was not associated with reduced activity, but rs2266780 was strongly associated with aberrant FMO3 mRNA splicing in both liver and brain (P⩽6.5 × 10-9). Surprisingly, in treatment-seeking European American smokers (n=1558) this allele was associated with reduced nicotine dependence, specifically with a longer time to first cigarette (P=9.0 × 10-4), but not with reduced cigarette consumption. As N-oxidation accounts for only a small percentage of hepatic nicotine metabolism we hypothesized that FMO3 genotype affects nicotine metabolism in the brain (unlike CYP2A6, FMO3 is expressed in human brain) or that nicotine-N-oxide itself has pharmacological activity. We demonstrate for the first time nicotine N-oxidation in human brain, mediated by FMO3 and FMO1, and show that nicotine-N-oxide modulates human α4β2 nicotinic receptor activity in vitro. These results indicate possible mechanisms for associations between FMO3 genotype and smoking behaviors, and suggest nicotine N-oxidation as a novel target to enhance smoking cessation.
Collapse
Affiliation(s)
- A M Teitelbaum
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - S E Murphy
- Department of Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - G Akk
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - T B Baker
- Department of Psychology, University of Wisconsin, Madison, WI, USA
| | - A Germann
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - L B von Weymarn
- Department of Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - L J Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - A Goate
- Neuroscience Genetics & Genomics Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - E D Kharasch
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - A J Bloom
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
14
|
Olfson E, Bloom J, Bertelsen S, Budde JP, Breslau N, Brooks A, Culverhouse R, Chan G, Chen LS, Chorlian D, Dick DM, Edenberg HJ, Hartz S, Hatsukami D, Hesselbrock VM, Johnson EO, Kramer JR, Kuperman S, Meyers JL, Nurnberger J, Porjesz B, Saccone NL, Schuckit MA, Stitzel J, Tischfield JA, Rice JP, Goate A, Bierut LJ. CYP2A6 metabolism in the development of smoking behaviors in young adults. Addict Biol 2018; 23:437-447. [PMID: 28032407 PMCID: PMC5491369 DOI: 10.1111/adb.12477] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/12/2016] [Accepted: 10/28/2016] [Indexed: 11/28/2022]
Abstract
Cytochrome P450 2A6 (CYP2A6) encodes the enzyme responsible for the majority of nicotine metabolism. Previous studies support that slow metabolizers smoke fewer cigarettes once nicotine dependent but provide conflicting results on the role of CYP2A6 in the development of dependence. By focusing on the critical period of young adulthood, this study examines the relationship of CYP2A6 variation and smoking milestones. A total of 1209 European American young adults enrolled in the Collaborative Study on the Genetics of Alcoholism were genotyped for CYP2A6 variants to calculate a previously well-validated metric that estimates nicotine metabolism. This metric was not associated with the transition from never smoking to smoking initiation nor with the transition from initiation to daily smoking (P > 0.4). But among young adults who had become daily smokers (n = 506), decreased metabolism was associated with increased risk of nicotine dependence (P = 0.03) (defined as Fagerström Test for Nicotine Dependence score ≥4). This finding was replicated in the Collaborative Genetic Study of Nicotine Dependence with 335 young adult daily smokers (P = 0.02). Secondary meta-analysis indicated that slow metabolizers had a 53 percent increased odds (OR = 1.53, 95 percent CI 1.11-2.11, P = 0.009) of developing nicotine dependence compared with normal metabolizers. Furthermore, secondary analyses examining four-level response of time to first cigarette after waking (>60, 31-60, 6-30, ≤5 minutes) demonstrated a robust effect of the metabolism metric in Collaborative Study on the Genetics of Alcoholism (P = 0.03) and Collaborative Genetic Study of Nicotine Dependence (P = 0.004), illustrating the important role of this measure of dependence. These findings highlight the complex role of CYP2A6 variation across different developmental stages of smoking behaviors.
Collapse
Affiliation(s)
- Emily Olfson
- Child Study Center and Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Joseph Bloom
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, USA
| | - Sarah Bertelsen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John P Budde
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Naomi Breslau
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Andrew Brooks
- Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Robert Culverhouse
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Grace Chan
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Li-Shiun Chen
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - David Chorlian
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Danielle M Dick
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sarah Hartz
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Dorothy Hatsukami
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, USA
| | - Victor M Hesselbrock
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Eric O Johnson
- Behavioral Health Epidemiology Program, RTI International, Research Triangle Park, NC, USA
| | - John R Kramer
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Samuel Kuperman
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jacquelyn L Meyers
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - John Nurnberger
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bernice Porjesz
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Nancy L Saccone
- Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | - Marc A Schuckit
- Department of Psychiatry, University of California, San Diego Medical School, San Diego, CA, USA
| | - Jerry Stitzel
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
| | - Jay A Tischfield
- Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - John P Rice
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Alison Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura J Bierut
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
15
|
Schuit E, Panagiotou OA, Munafò MR, Bennett DA, Bergen AW, David SP, Cochrane Tobacco Addiction Group. Pharmacotherapy for smoking cessation: effects by subgroup defined by genetically informed biomarkers. Cochrane Database Syst Rev 2017; 9:CD011823. [PMID: 28884473 PMCID: PMC6483659 DOI: 10.1002/14651858.cd011823.pub2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Smoking cessation therapies are not effective for all smokers, and researchers are interested in identifying those subgroups of individuals (e.g. based on genotype) who respond best to specific treatments. OBJECTIVES To assess whether quit rates vary by genetically informed biomarkers within pharmacotherapy treatment arms and as compared with placebo. To assess the effects of pharmacotherapies for smoking cessation in subgroups of smokers defined by genotype for identified genome-wide significant polymorphisms. SEARCH METHODS We searched the Cochrane Tobacco Addiction Group specialised register, clinical trial registries, and genetics databases for trials of pharmacotherapies for smoking cessation from inception until 16 August 2016. SELECTION CRITERIA We included randomised controlled trials (RCTs) that recruited adult smokers and reported pharmacogenomic analyses from trials of smoking cessation pharmacotherapies versus controls. Eligible trials included those with data on a priori genome-wide significant (P < 5 × 10-8) single-nucleotide polymorphisms (SNPs), replicated non-SNPs, and/or the nicotine metabolite ratio (NMR), hereafter collectively described as biomarkers. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. The primary outcome was smoking abstinence at six months after treatment. The secondary outcome was abstinence at end of treatment (EOT). We conducted two types of meta-analyses- one in which we assessed smoking cessation of active treatment versus placebo within genotype groups, and another in which we compared smoking cessation across genotype groups within treatment arms. We carried out analyses separately in non-Hispanic whites (NHWs) and non-Hispanic blacks (NHBs). We assessed heterogeneity between genotype groups using T², I², and Cochrane Q statistics. MAIN RESULTS Analyses included 18 trials including 9017 participants, of whom 6924 were NHW and 2093 NHB participants. Data were available for the following biomarkers: nine SNPs (rs1051730 (CHRNA3); rs16969968, rs588765, and rs2036527 (CHRNA5); rs3733829 and rs7937 (in EGLN2, near CYP2A6); rs1329650 and rs1028936 (LOC100188947); and rs215605 (PDE1C)), two variable number tandem repeats (VNTRs; DRD4 and SLC6A4), and the NMR. Included data produced a total of 40 active versus placebo comparisons, 16 active versus active comparisons, and 64 between-genotype comparisons within treatment arms.For those meta-analyses showing statistically significant heterogeneity between genotype groups, we found the quality of evidence (GRADE) to be generally moderate. We downgraded quality most often because of imprecision or risk of bias due to potential selection bias in genotyping trial participants. Comparisons of relative treatment effects by genotypeFor six-month abstinence, we found statistically significant heterogeneity between genotypes (rs16969968) for nicotine replacement therapy (NRT) versus placebo at six months for NHB participants (P = 0.03; n = 2 trials), but not for other biomarkers or treatment comparisons. Six-month abstinence was increased in the active NRT group as compared to placebo among participants with a GG genotype (risk ratio (RR) 1.47, 95% confidence interval (CI) 1.07 to 2.03), but not in the combined group of participants with a GA or AA genotype (RR 0.43, 95% CI 0.15 to 1.26; ratio of risk ratios (RRR) GG vs GA or AA of 3.51, 95% CI 1.19 to 10.3). Comparisons of treatment effects between genotype groups within pharmacotherapy randomisation armsFor those receiving active NRT, treatment was more effective in achieving six-month abstinence among individuals with a slow NMR than among those with a normal NMR among NHW and NHB combined participants (normal NMR vs slow NMR: RR 0.54, 95% CI 0.37 to 0.78; n = 2 trials). We found no such differences in treatment effects between genotypes at six months for any of the other biomarkers among individuals who received pharmacotherapy or placebo. AUTHORS' CONCLUSIONS We did not identify widespread differential treatment effects of pharmacotherapy based on genotype. Some genotype groups within certain ethnic groups may benefit more from NRT or may benefit less from the combination of bupropion with NRT. The reader should interpret these results with caution because none of the statistically significant meta-analyses included more than two trials per genotype comparison, many confidence intervals were wide, and the quality of this evidence (GRADE) was generally moderate. Although we found evidence of superior NRT efficacy for NMR slow versus normal metabolisers, because of the lack of heterogeneity between NMR groups, we cannot conclude that NRT is more effective for slow metabolisers. Access to additional data from multiple trials is needed, particularly for comparisons of different pharmacotherapies.
Collapse
Affiliation(s)
- Ewoud Schuit
- Stanford UniversityMeta‐Research Innovation Center at Stanford (METRICS)StanfordCAUSA
- University Medical Center UtrechtCochrane NetherlandsUtrechtNetherlands
- University Medical Center UtrechtJulius Center for Health Sciences and Primary CareUtrechtNetherlands
| | - Orestis A. Panagiotou
- School of Public Health, Brown UniversityDepartment of Health Services, Policy & Practice121 S. Main StreetProvidenceRIUSA02903
| | - Marcus R Munafò
- University of BristolSchool of Experimental Psychology and MRC Integrative Epidemiology Unit8 Woodland RoadBristolUKBS8 1TN
| | - Derrick A Bennett
- University of OxfordClinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population HealthRichard Doll BuildingOld Road CampusOxfordUKOX3 7LF
| | | | - Sean P David
- Stanford UniversityDivision of Primary Care and Population Health, Department of MedicineStanfordCaliforniaUSA94304‐5559
| | | |
Collapse
|
16
|
Yuan JM, Nelson HH, Carmella SG, Wang R, Kuriger-Laber J, Jin A, Adams-Haduch J, Hecht SS, Koh WP, Murphy SE. CYP2A6 genetic polymorphisms and biomarkers of tobacco smoke constituents in relation to risk of lung cancer in the Singapore Chinese Health Study. Carcinogenesis 2017; 38:411-418. [PMID: 28182203 PMCID: PMC6248819 DOI: 10.1093/carcin/bgx012] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/16/2017] [Accepted: 01/24/2017] [Indexed: 01/01/2023] Open
Abstract
Cytochrome P450 2A6 (CYP2A6) catalyzes the metabolism of nicotine and the tobacco-specific lung carcinogen, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). Genetic variation in CYP2A6 may affect smoking behavior and contribute to lung cancer risk. A nested case-control study of 197 lung cancer cases and 197 matched controls was conducted within a prospective cohort of 63 257 Chinese men and women in Singapore. Quantified were five genetic variants of CYP2A6 (*1A, *4, *7, *9 and *12) and urinary metabolites of nicotine [total nicotine, total cotinine, total trans-3'-hydroxycotinine (3HC)] and NNK (total NNAL, free NNAL, NNAL-glucuronide, NNAL-N-glucuronide, and NNAL-O-glucuronide). Higher urinary metabolites of nicotine and NNK were significantly associated with a 2- to 3-fold increased risk of lung cancer after adjustment for smoking intensity and duration. Lower CYP2A6-determined nicotine metabolizer status was significantly associated with a lower ratio of total 3HC over total cotinine, lower total nicotine equivalent and reduced risk of developing lung cancer (all Ptrend < 0.001). Compared with normal metabolizers, odds ratios (95% confidence intervals) of developing lung cancer for intermediate, slow and poor metabolizers determined by CYP2A6 genotypes were 0.85 (0.41-1.77), 0.55 (0.28-1.08) and 0.32 (0.15-0.70), respectively, after adjustment for smoking intensity and duration and urinary total nicotine equivalents. Thus the reduced risk of lung cancer in smokers with lower CYP2A6 activity may be explained by lower consumption of cigarettes, less intense smoking and reduced CYP2A6-catalyzed activation of the tobacco-specific lung carcinogen NNK.
Collapse
Affiliation(s)
- Jian-Min Yuan
- Division of Cancer Control and Population Sciences, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Heather H Nelson
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Steven G Carmella
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Renwei Wang
- Division of Cancer Control and Population Sciences, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | | | - Aizhen Jin
- National Registry of Diseases Office, Health Promotion Board, Singapore, Singapore
| | - Jennifer Adams-Haduch
- Division of Cancer Control and Population Sciences, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Stephen S Hecht
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Woon-Puay Koh
- Duke-NUS Medical School Singapore, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore and
| | - Sharon E Murphy
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and BioPhysics, University of Minnesota, MN, USA
| |
Collapse
|
17
|
Richmond-Rakerd LS, Otto JM, Slutske WS, Ehlers CL, Wilhelmsen KC, Gizer IR. A Novel Tobacco Use Phenotype Suggests the 15q25 and 19q13 Loci May be Differentially Associated With Cigarettes per Day and Tobacco-Related Problems. Nicotine Tob Res 2017; 19:426-434. [PMID: 27663783 PMCID: PMC5968625 DOI: 10.1093/ntr/ntw260] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 09/22/2016] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Tobacco use is associated with variation at the 15q25 gene cluster and the cytochrome P450 (CYP) genes CYP2A6 and CYP2B6. Despite the variety of outcomes associated with these genes, few studies have adopted a data-driven approach to defining tobacco use phenotypes for genetic association analyses. We used factor analysis to generate a tobacco use measure, explored its incremental validity over a simple indicator of tobacco involvement: cigarettes per day (CPD), and tested both phenotypes in a genetic association study. METHODS Data were from the University of California, San Francisco Family Alcoholism Study (n = 1942) and a Native American sample (n = 255). Factor analyses employed a broad array of tobacco use variables to establish the candidate phenotype. Subsequently, we conducted tests for association with variants in the nicotinic acetylcholine receptor and CYP genes. We explored associations with CPD and our measure. We then examined whether the variants most strongly associated with our measure remained associated after controlling for CPD. RESULTS Analyses identified one factor that captured tobacco-related problems. Variants at 15q25 were significantly associated with CPD after multiple testing correction (rs938682: p = .00002, rs1051730: p = .0003, rs16969968: p = .0003). No significant associations were obtained with the tobacco use phenotype; however, suggestive associations were observed for variants in CYP2B6 near CYP2A6 (rs45482602: ps = .0082, .0075) and CYP4Z2P (rs10749865: ps = .0098, .0079). CONCLUSIONS CPD captures variation at 15q25. Although strong conclusions cannot be drawn, these finding suggest measuring additional dimensions of problems may detect genetic variation not accounted for by smoking quantity. Replication in independent samples will help further refine phenotype definition efforts. IMPLICATIONS Different facets of tobacco-related problems may index unique genetic risk. CPD, a simple measure of tobacco consumption, is associated with variants at the 15q25 gene cluster. Additional dimensions of tobacco problems may help to capture variation at 19q13. Results demonstrate the utility of adopting a data-driven approach to defining phenotypes for genetic association studies of tobacco involvement and provide results that can inform replication efforts.
Collapse
Affiliation(s)
- Leah S Richmond-Rakerd
- Department of Psychological Sciences, University of Missouri, Columbia, MO
- Alcoholism Research Center at Washington University School of Medicine, St. Louis, MO
| | - Jacqueline M Otto
- Department of Psychological Sciences, University of Missouri, Columbia, MO
- Alcoholism Research Center at Washington University School of Medicine, St. Louis, MO
| | - Wendy S Slutske
- Department of Psychological Sciences, University of Missouri, Columbia, MO
- Alcoholism Research Center at Washington University School of Medicine, St. Louis, MO
| | - Cindy L Ehlers
- Department of Molecular and Cellular Neurosciences (CLE), The Scripps Research Institute, La Jolla, CA
| | - Kirk C Wilhelmsen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ian R Gizer
- Department of Psychological Sciences, University of Missouri, Columbia, MO
- Alcoholism Research Center at Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
18
|
Baurley JW, Edlund CK, Pardamean CI, Conti DV, Bergen AW. Smokescreen: a targeted genotyping array for addiction research. BMC Genomics 2016; 17:145. [PMID: 26921259 PMCID: PMC4769529 DOI: 10.1186/s12864-016-2495-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 02/17/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Addictive disorders are a class of chronic, relapsing mental disorders that are responsible for increased risk of mental and medical disorders and represent the largest, potentially modifiable cause of death. Tobacco dependence is associated with increased risk of disease and premature death. While tobacco control efforts and therapeutic interventions have made good progress in reducing smoking prevalence, challenges remain in optimizing their effectiveness based on patient characteristics, including genetic variation. In order to maximize collaborative efforts to advance addiction research, we have developed a genotyping array called Smokescreen. This custom array builds upon previous work in the analyses of human genetic variation, the genetics of addiction, drug metabolism, and response to therapy, with an emphasis on smoking and nicotine addiction. RESULTS The Smokescreen genotyping array includes 646,247 markers in 23 categories. The array design covers genome-wide common variation (65.67, 82.37, and 90.72% in African (YRI), East Asian (ASN), and European (EUR) respectively); most of the variation with a minor allele frequency ≥ 0.01 in 1014 addiction genes (85.16, 89.51, and 90.49% for YRI, ASN, and EUR respectively); and nearly all variation from the 1000 Genomes Project Phase 1, NHLBI GO Exome Sequencing Project and HapMap databases in the regions related to smoking behavior and nicotine metabolism: CHRNA5-CHRNA3-CHRNB4 and CYP2A6-CYP2B6. Of the 636 pilot DNA samples derived from blood or cell line biospecimens that were genotyped on the array, 622 (97.80%) passed quality control. In passing samples, 90.08% of markers passed quality control. The genotype reproducibility in 25 replicate pairs was 99.94%. For 137 samples that overlapped with HapMap2 release 24, the genotype concordance was 99.76%. In a genome-wide association analysis of the nicotine metabolite ratio in 315 individuals participating in nicotine metabolism laboratory studies, we identified genome-wide significant variants in the CYP2A6 region (min p = 9.10E-15). CONCLUSIONS We developed a comprehensive genotyping array for addiction research and demonstrated its analytic validity and utility through pilot genotyping of HapMap and study samples. This array allows researchers to perform genome-wide, candidate gene, and pathway-based association analyses of addiction, tobacco-use, treatment response, comorbidities, and associated diseases in a standardized, high-throughput platform.
Collapse
Affiliation(s)
- James W Baurley
- BioRealm LLC, 6101 W. Centinela Ave., Suite 270, Culver City, CA, 90230-6359, USA.
| | - Christopher K Edlund
- BioRealm LLC, 6101 W. Centinela Ave., Suite 270, Culver City, CA, 90230-6359, USA.
| | - Carissa I Pardamean
- BioRealm LLC, 6101 W. Centinela Ave., Suite 270, Culver City, CA, 90230-6359, USA.
| | - David V Conti
- BioRealm LLC, 6101 W. Centinela Ave., Suite 270, Culver City, CA, 90230-6359, USA.
| | - Andrew W Bergen
- BioRealm LLC, 6101 W. Centinela Ave., Suite 270, Culver City, CA, 90230-6359, USA.
| |
Collapse
|
19
|
Park SL, Tiirikainen MI, Patel YM, Wilkens LR, Stram DO, Le Marchand L, Murphy SE. Genetic determinants of CYP2A6 activity across racial/ethnic groups with different risks of lung cancer and effect on their smoking intensity. Carcinogenesis 2016; 37:269-279. [PMID: 26818358 DOI: 10.1093/carcin/bgw012] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/19/2016] [Indexed: 01/22/2023] Open
Abstract
Genetic variation in cytochrome P450 2A6 (CYP2A6) gene is the primary contributor to the intraindividual and interindividual differences in nicotine metabolism and has been found to influence smoking intensity. However, no study has evaluated the relationship between CYP2A6 genetic variants and the CYP2A6 activity ratio (total 3-hydroxycotinine/cotinine) and their influence on smoking intensity [total nicotine equivalents (TNE)], across five racial/ethnic groups found to have disparate rates of lung cancer. This study genotyped 10 known functional CYP2A6 genetic or copy number variants in 2115 current smokers from the multiethnic cohort study [African Americans (AA) = 350, Native Hawaiians (NH) = 288, Whites = 413, Latinos (LA) = 437 and Japanese Americans (JA) = 627] to conduct such an investigation. Here, we found that LA had the highest CYP2A6 activity followed by Whites, AA, NH and JA, who had the lowest levels. Adjusting for age, sex, race/ethnicity and body mass index, we found that CYP2A6 diplotypes were predictive of TNE levels, particularly in AA and JA (P trend < 0.0001). However, only in JA did the association remain after accounting for cigarettes per day. Also, it is only in this population that the lower activity ratio supports lower TNE levels, carcinogen exposure and thereby lower risk of lung cancer. Despite the association between nicotine metabolism (CYP2A6 activity phenotype and diplotypes) and smoking intensity (TNE), CYP2A6 levels did not correlate with the higher TNE levels found in AA nor the lower TNE levels found in LA, suggesting that other factors may influence smoking dose in these populations. Therefore, further study in these populations is recommended.
Collapse
Affiliation(s)
| | - Maarit I Tiirikainen
- Epidemiology Program, University of Hawaii Cancer Center , Honolulu, HI 96813 , USA and
| | | | - Lynne R Wilkens
- Epidemiology Program, University of Hawaii Cancer Center , Honolulu, HI 96813 , USA and
| | | | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center , Honolulu, HI 96813 , USA and
| | - Sharon E Murphy
- Department of Biochemistry, Molecular Biology and Biophysics, Masonic Cancer Center, University of Minnesota , 2231 6th Street SE, 2-127 CCRB, Minneapolis, MN 55455 , USA
| |
Collapse
|
20
|
Cannon DS, Mermelstein RJ, Medina TR, Pugach O, Hedeker D, Weiss RB. CYP2A6 Effects on Subjective Reactions to Initial Smoking Attempt. Nicotine Tob Res 2015; 18:637-41. [PMID: 26541911 DOI: 10.1093/ntr/ntv244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/15/2015] [Indexed: 12/11/2022]
Abstract
INTRODUCTION In very novice smokers, CYP2A6 genotypes that reduce nicotine metabolism to an intermediate rate may increase smoking risk, relative to both normal and slow rates. The present study examined the hypothesis that intermediate metabolism variants are associated with greater pleasurable effects of the initial smoking attempt than either normal or slow metabolism variants. METHODS Participants were novice smokers (N = 261, 65% female) of European descent. Predicted nicotine metabolic rate based on CYP2A6 diplotypes (CYP2A6 Diplotype Predicted Rate [CDPR]) was partitioned into Normal, Intermediate, and Slow categories using a metabolism metric. Subjective reactions to the initial smoking attempt were assessed by the Pleasurable Smoking Experiences (PSE) scale, which was collected within 3 years of the initial smoking attempt. The effect of CDPR on PSE was tested using a generalized linear model in which CDPR was dummy coded and Intermediate CDPR was the reference condition. Gender was included in the model as a control for higher PSE scores by males. RESULTS Lower PSE scores were associated with Normal CDPR, β = -0.34, P = .008, and Slow CDPR, β = -0.52, P = .001, relative to Intermediate CDPR. CONCLUSIONS Intermediate CDPR-enhanced pleasurable effects of the initial smoking attempt relative to other CYP2A6 variants. This finding is consistent with the hypothesis that the risk effect of Intermediate CDPR on early smoking is a function of optimal pleasurable effects. IMPLICATIONS This study supports our recent hypothesis that CYP2A6 diplotypes that encode intermediate nicotine metabolism rate are associated with enhanced pleasurable events following the initial smoking attempt, compared with diplotypes that encode either normal or slow metabolism. This hypothesis was offered to account for our unexpected previous finding of enhanced smoking risk in very novice smokers associated with intermediate metabolism rate. Our new finding encourages further investigation of time-dependent relations between CYP2A6 effects and smoking motives, and it encourages laboratory study of the mechanisms underlying the initial smoking enhancement in novice smokers associated with intermediate metabolism.
Collapse
Affiliation(s)
- Dale S Cannon
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT;
| | - Robin J Mermelstein
- Institute for Health Research and Policy and Psychology Department, University of Illinois, Chicago, IL
| | - Tait R Medina
- Institute for Public Health Research and Policy, University of Illinois, Chicago, IL
| | - Oksana Pugach
- Institute for Public Health Research and Policy, University of Illinois, Chicago, IL
| | - Donald Hedeker
- Department of Health Sciences, University of Chicago, Chicago, IL
| | - Robert B Weiss
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT
| |
Collapse
|
21
|
Abstract
For the past 30 years, research examining predictors of successful smoking cessation treatment response has focused primarily on clinical variables, such as levels of tobacco dependence, craving, and self-efficacy. However, recent research has begun to determine biomarkers (such as genotype, nicotine and metabolite levels, and brain imaging findings) that may have utility in predicting smoking cessation. For genotype, genes associated with nicotinic acetylcholine receptors (nAChRs) and related proteins have been found to predict response to first-line medications (e.g. nicotine replacement therapy [NRT], bupropion, or varenicline) or quitting over time without a controlled treatment trial. For nicotine and metabolite levels, function of the cytochrome P450 2A6 liver enzyme, which can be assessed with the nicotine metabolite ratio or via genotype, has been found to predict response, with slow nicotine metabolizers having less severe nicotine dependence and a greater likelihood of quitting with NRT than normal metabolizers. For brain imaging, decreased activation of brain regions associated with emotion regulation and increased connectivity in emotion regulation networks, increased responsiveness to pleasant cues, and altered activation with the Stroop effect have been found in smokers who quit with the first-line medications listed above or counseling. In addition, our group recently demonstrated that lower pre-treatment brain nAChR density is associated with a greater chance of quitting smoking with NRT or placebo. Several of these studies found that specific biomarkers may provide additional information for predicting response beyond subjective symptom or rating scale measures, thereby giving an initial indication that biomarkers may, in the future, be useful for guiding smoking cessation treatment intensity, duration, and type.
Collapse
|
22
|
Cannon DS, Medina TR, Mermelstein RJ, Hedeker D, Bakian AV, Coon H, Cook EH, Hamil C, Weiss RB. CYP2A6 Longitudinal Effects in Young Smokers. Nicotine Tob Res 2015; 18:196-203. [PMID: 25744963 DOI: 10.1093/ntr/ntv049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 02/16/2015] [Indexed: 11/13/2022]
Abstract
INTRODUCTION The present study sought to identify time-dependent within-participant effects of CYP2A6 genotypes on smoking frequency and nicotine dependence in young smokers. METHODS Predicted nicotine metabolic rate based on CYP2A6 diplotypes (CYP2A6 diplotype predicted rate [CDPR]) was partitioned into Normal, Intermediate, and Slow categories using a metabolism metric. Growth-curve models characterized baseline and longitudinal CDPR effects with data from eight longitudinal assessments during a 6-year period (from approximately age 16-22) in young smokers of European descent (N = 296, 57% female) who had smoked less than 100 cigarettes lifetime at baseline and more than that amount by Year 6. Phenotypes were number of days smoked during the previous 30 days and a youth version of the Nicotine Dependence Syndrome Scale (NDSS). A zero-inflated Poisson growth-curve model was used to account for the preponderance of zero days smoked. RESULTS At baseline, Intermediate CDPR was a risk factor relative to both Normal and Slow CDPR for smoking frequency and the NDSS. Slow CDPR was associated with the highest probability of smoking discontinuation at baseline. However, due to CDPR time trend differences, by young adulthood these baseline effects had been reordered such that the greatest risks for smoking frequency and the NDSS were associated with Normal CDPR. CONCLUSIONS Reduced metabolism CYP2A6 genotypes are associated with both risk and protective effects in novice smokers. However, differences in the time-by-CDPR effects result in a reordering of genotype effects such that normal metabolism becomes the risk variant by young adulthood, as has been reliably reported in older smokers.
Collapse
Affiliation(s)
- Dale S Cannon
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT;
| | - Tait R Medina
- Department of Sociology, Indiana University, Bloomington, IN; Institute for Health Research and Policy, University of Illinois, Chicago, IL
| | - Robin J Mermelstein
- Institute for Health Research and Policy and Psychology Department, University of Illinois, Chicago, IL
| | - Donald Hedeker
- Department of Health Sciences, University of Chicago, Chicago, IL
| | - Amanda V Bakian
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT
| | - Hilary Coon
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT
| | - Edwin H Cook
- Institute for Juvenile Research, Department of Psychiatry, University of Illinois, Chicago, IL
| | - Cindy Hamil
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT
| | - Robert B Weiss
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT
| |
Collapse
|
23
|
Murphy SE, Park SSL, Thompson EF, Wilkens LR, Patel Y, Stram DO, Le Marchand L. Nicotine N-glucuronidation relative to N-oxidation and C-oxidation and UGT2B10 genotype in five ethnic/racial groups. Carcinogenesis 2014; 35:2526-33. [PMID: 25233931 PMCID: PMC4216060 DOI: 10.1093/carcin/bgu191] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 08/27/2014] [Accepted: 09/04/2014] [Indexed: 12/16/2022] Open
Abstract
Nicotine metabolism influences smoking behavior and differences in metabolism probably contribute to ethnic variability in lung cancer risk. We report here on the proportion of nicotine metabolism by cytochrome P450 2A6-catalyzed C-oxidation, UDP-glucuronosyl transferase 2B10 (UGT2B10)-catalyzed N-glucuronidation and flavin monooxygenase 3-catalyzed N-oxidation in five ethnic/racial groups and the role of UGT2B10 genotype on the metabolic patterns observed. Nicotine and its metabolites were quantified in urine from African American (AA, n = 364), Native Hawaiian (NH, n = 311), White (n = 437), Latino (LA, n = 453) and Japanese American (JA, n = 674) smokers. Total nicotine equivalents, the sum of nicotine and six metabolites, and nicotine metabolism phenotypes were calculated. The relationship of UGT2B10 genotype to nicotine metabolic pathways was determined for each group; geometric means were computed and adjusted for age, sex, creatinine, and body mass index. Nicotine metabolism patterns were unique across the groups, C-oxidation was lowest in JA and NH (P < 0.0001), and N-glucuronidation lowest in AA (P < 0.0001). There was no difference in C-oxidation among Whites and AA and LA. Nicotine and cotinine glucuronide ratios were 2- and 3-fold lower in AA compared with Whites. Two UGT variants, a missense mutation (Asp67Tyr, rs61750900) and a splice variant (rs116294140) accounted for 33% of the variation in glucuronidation. In AA, the splice variant accounted for the majority of the reduced nicotine glucuronidation. UGT2B10 variant allele carriers had increased levels of C-oxidation (P = 0.0099). Our data indicate that the relative importance of nicotine metabolic pathways varies by ethnicity, and all pathways should be considered when characterizing the role of nicotine metabolism on smoking behavior and cancer risk.
Collapse
Affiliation(s)
- Sharon E Murphy
- Department of Biochemistry Molecular Biology and Biophysics and Masonic Cancer Center, University of Minnesota, 2-127 CCRB, 2231 6th Street SE, Minneapolis, MN 55455, USA, Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA and Cancer Research Center of Hawai'i, University of Hawai'i, Honolulu, HI 96813, USA
| | - Sung-Shim L Park
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA and
| | - Elizabeth F Thompson
- Department of Biochemistry Molecular Biology and Biophysics and Masonic Cancer Center, University of Minnesota, 2-127 CCRB, 2231 6th Street SE, Minneapolis, MN 55455, USA, Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA and Cancer Research Center of Hawai'i, University of Hawai'i, Honolulu, HI 96813, USA
| | - Lynne R Wilkens
- Cancer Research Center of Hawai'i, University of Hawai'i, Honolulu, HI 96813, USA
| | - Yesha Patel
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA and
| | - Daniel O Stram
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA and
| | - Loic Le Marchand
- Cancer Research Center of Hawai'i, University of Hawai'i, Honolulu, HI 96813, USA
| |
Collapse
|
24
|
Bloom AJ, Hartz SM, Baker TB, Chen LS, Piper ME, Fox L, Martinez M, Hatsukami D, Johnson EO, Laurie CC, Saccone NL, Goate A, Bierut LJ. Beyond cigarettes per day. A genome-wide association study of the biomarker carbon monoxide. Ann Am Thorac Soc 2014; 11:1003-10. [PMID: 25072098 PMCID: PMC4214060 DOI: 10.1513/annalsats.201401-010oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 06/04/2014] [Indexed: 12/19/2022] Open
Abstract
RATIONALE The CHRNA5-CHRNA3-CHRNB4 locus is associated with self-reported smoking behavior and also harbors the strongest genetic associations with chronic obstructive pulmonary disease (COPD) and lung cancer. Because the associations with lung disease remain after adjustment for self-reported smoking behaviors, it has been asserted that CHRNA5-CHRNA3-CHRNB4 variants increase COPD and lung cancer susceptibility independently of their effects on smoking. OBJECTIVES To compare the genetic associations of exhaled carbon monoxide (CO), a biomarker of current cigarette exposure, with self-reported smoking behaviors. METHODS A total of 1,521 European American and 247 African American current smokers recruited into smoking cessation studies were assessed for CO at intake before smoking cessation. DNA samples were genotyped using the Illumina Omni2.5 microarray. Genetic associations with CO and smoking behaviors (cigarettes smoked per day, Fagerstrom test for nicotine dependence) were studied. MEASUREMENTS AND MAIN RESULTS Variants in the CHRNA5-CHRNA3-CHRNB4 locus, including rs16969968, a nonsynonymous variant in CHRNA5, are genomewide association study-significantly associated with CO (β = 2.66; 95% confidence interval [CI], 1.74-3.58; P = 1.65 × 10(-8)), and this association remains strong after adjusting for smoking behavior (β = 2.18; 95% CI, 1.32-3.04; P = 7.47 × 10(-7)). The correlation between CO and cigarettes per day is statistically significantly lower (z = 3.43; P = 6.07 × 10(-4)) in African Americans (r = 0.14; 95% CI, 0.02-0.26; P = 0.003) than in European-Americans (r = 0.36; 95% CI, 0.31-0.40; P = 0.0001). CONCLUSIONS Exhaled CO, a biomarker that is simple to measure, captures aspects of cigarette smoke exposure in current smokers beyond the number of cigarettes smoked per day. Behavioral measures of smoking are therefore insufficient indices of cigarette smoke exposure, suggesting that genetic associations with COPD or lung cancer that persist after adjusting for self-reported smoking behavior may still reflect genetic effects on smoking exposure.
Collapse
|
25
|
Novel CYP2A6 variants identified in African Americans are associated with slow nicotine metabolism in vitro and in vivo. Pharmacogenet Genomics 2014; 24:118-28. [PMID: 24305170 DOI: 10.1097/fpc.0000000000000026] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Nicotine, the main addictive ingredient in tobacco, is metabolically inactivated to cotinine primarily by the hepatic enzyme CYP2A6. Considerable genetic variation in the CYP2A6 gene results in large variation in the rates of nicotine metabolism, which in turn alters smoking behaviours (e.g. amount of cigarettes smoked, risk for dependence and success in smoking cessation). The aim of this study was to identify and characterize novel variants in CYP2A6. MATERIALS AND METHODS The CYP2A6 gene from African American phenotypically slow nicotine metabolizers was sequenced and seven novel variants were identified [CYP2A6*39 (V68M), CYP2A6*40 (I149M), CYP2A6*41 (R265Q), CYP2A6*42 (I268T), CYP2A6*43 (T303I), CYP2A6*44 (E390K), CYP2A6*44 (L462P)]. Variants were introduced into a bi-cistronic cDNA expression construct containing CYP2A6 and P450 oxidoreductase and assessed for protein expression, enzymatic activity and stability as evaluated using western blotting and nicotine metabolism. Genotyping assays were developed and allelic frequencies were assessed in 534 African Americans. RESULTS The variants showed significantly lower protein expression (P<0.001) when compared with the wild-type as well as reduced metabolism of nicotine to cotinine when controlling for cDNA expression using P450 oxidoreductase (P<0.001). The variants also showed reduced stability at 37°C. Allelic frequencies ranged from 0.1 to 0.6% with a collective genotype frequency of 3.2%; the impact in vitro correlated significantly with in-vivo activity (R(2)=0.40-0.48, P<0.05). Together, those with a novel variant had significantly lower nicotine metabolism in vivo than those without genetic variants (P<0.01). CONCLUSION Here, we identified a number of novel variants with reduced/loss of CYP2A6 activity, increasing our understanding of CYP2A6 genetic variability.
Collapse
|
26
|
Cho MH, McDonald MLN, Zhou X, Mattheisen M, Castaldi PJ, Hersh CP, Demeo DL, Sylvia JS, Ziniti J, Laird NM, Lange C, Litonjua AA, Sparrow D, Casaburi R, Barr RG, Regan EA, Make BJ, Hokanson JE, Lutz S, Dudenkov TM, Farzadegan H, Hetmanski JB, Tal-Singer R, Lomas DA, Bakke P, Gulsvik A, Crapo JD, Silverman EK, Beaty TH. Risk loci for chronic obstructive pulmonary disease: a genome-wide association study and meta-analysis. THE LANCET. RESPIRATORY MEDICINE 2014; 2:214-25. [PMID: 24621683 PMCID: PMC4176924 DOI: 10.1016/s2213-2600(14)70002-5] [Citation(s) in RCA: 257] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND The genetic risk factors for susceptibility to chronic obstructive pulmonary disease (COPD) are still largely unknown. Additional genetic variants are likely to be identified by genome-wide association studies in larger cohorts or specific subgroups. We sought to identify risk loci for moderate to severe and severe COPD with data from several cohort studies. METHODS We combined genome-wide association analysis data from participants in the COPDGene study (non-Hispanic white and African-American ethnic origin) and the ECLIPSE, NETT/NAS, and Norway GenKOLS studies (self-described white ethnic origin). We did analyses comparing control individuals with individuals with moderate to severe COPD and with a subset of individuals with severe COPD. Single nucleotide polymorphisms yielding a p value of less than 5 × 10(-7) in the meta-analysis at loci not previously described were genotyped in individuals from the family-based ICGN study. We combined results in a joint meta-analysis (threshold for significance p<5 × 10(-8)). FINDINGS Analysis of 6633 individuals with moderate to severe COPD and 5704 control individuals confirmed association at three known loci: CHRNA3 (p=6·38 × 10(-14)), FAM13A (p=1·12 × 10(-14)), and HHIP (p=1·57 × 10(-12)). We also showed significant evidence of association at a novel locus near RIN3 (p=5·25 × 10(-9)). In the overall meta-analysis (ie, including data from 2859 ICGN participants), the association with RIN3 remained significant (p=5·4 × 10(-9)). 3497 individuals were included in our analysis of severe COPD. The effect estimates for the loci near HHIP and CHRNA3 were significantly stronger in severe disease than in moderate to severe disease (p<0·01). We also identified associations at two additional loci: MMP12 (overall joint meta-analysis p=2·6 × 10(-9)) and TGFB2 (overall joint meta-analysis p=8·3 × 10(-9)). INTERPRETATION We have confirmed associations with COPD at three known loci and identified three new genome-wide significant associations. Genetic variants other than in α-1 antitrypsin increase the risk of COPD. FUNDING US National Heart, Lung, and Blood Institute; the Alpha-1 Foundation; the COPD Foundation through contributions from AstraZeneca, Boehringer Ingelheim, Novartis, and Sepracor; GlaxoSmithKline; Centers for Medicare and Medicaid Services; Agency for Healthcare Research and Quality; and US Department of Veterans Affairs.
Collapse
Affiliation(s)
- Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| | - Merry-Lynn N McDonald
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Manuel Mattheisen
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard School of Public Health, Boston, MA, USA
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Dawn L Demeo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jody S Sylvia
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - John Ziniti
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Nan M Laird
- Harvard School of Public Health, Boston, MA, USA
| | | | - Augusto A Litonjua
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - David Sparrow
- School of Public Health and School of Medicine, Boston University, Boston, MA, USA; Veterans Administration Boston Healthcare System, Boston, MA, USA
| | - Richard Casaburi
- Los Angeles Biomedical Research Institute at Harbor UCLA Medical Center, Torrance, CA, USA
| | - R Graham Barr
- Department of Medicine, College of Physicians and Surgeons, Mailman School of Public Health, Columbia University, New York, NY, USA; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Elizabeth A Regan
- National Jewish Health, Denver, CO, USA; Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, USA
| | | | - John E Hokanson
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, USA
| | - Sharon Lutz
- Department of Bioinformatics and Statistics, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, USA
| | - Tanda Murray Dudenkov
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Homayoon Farzadegan
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Jacqueline B Hetmanski
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Ruth Tal-Singer
- GlaxoSmithKline Research and Development, King Of Prussia, PA, USA
| | | | - Per Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Amund Gulsvik
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | | | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Terri H Beaty
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
27
|
Abstract
Addictions are prevalent psychiatric disorders that confer remarkable personal and social burden. Despite substantial evidence for their moderate, yet robust, heritability (approx. 50%), specific genetic mechanisms underlying their development and maintenance remain unclear. The goal of this selective review is to highlight progress in unveiling the genetic underpinnings of addiction. First, we revisit the basis for heritable variation in addiction before reviewing the most replicable candidate gene findings and emerging signals from genomewide association studies for alcohol, nicotine and cannabis addictions. Second, we survey the modest but growing field of neurogenetics examining how genetic variation influences corticostriatal structure, function, and connectivity to identify neural mechanisms that may underlie associations between genetic variation and addiction. Third, we outline how extant genomic findings are being used to develop and refine pharmacotherapies. Finally, as sample sizes for genetically informed studies of addiction approach critical mass, we posit five exciting possibilities that may propel further discovery (improved phenotyping, rare variant discovery, gene-environment interplay, epigenetics, and novel neuroimaging designs).
Collapse
|
28
|
Chen LS, Bloom AJ, Baker TB, Smith SS, Piper ME, Martinez M, Saccone N, Hatsukami D, Goate A, Bierut L. Pharmacotherapy effects on smoking cessation vary with nicotine metabolism gene (CYP2A6). Addiction 2014; 109:128-137. [PMID: 24033696 PMCID: PMC3946972 DOI: 10.1111/add.12353] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 06/27/2013] [Accepted: 09/03/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND AIMS Evidence suggests that both the nicotinic receptor α5 subunit (CHRNA5) and Cytochrome P450 2A6 (CYP2A6) genotypes influence smoking cessation success and response to pharmacotherapy. We examine the effect of CYP2A6 genotype on smoking cessation success and response to cessation pharmacotherapy, and combine these effects with those of CHRNA5 genotypes. DESIGN Placebo-controlled randomized smoking cessation trial. SETTING Ambulatory care facility in Wisconsin, USA. PARTICIPANTS Smokers (n = 709) of European ancestry were randomized to placebo, bupropion, nicotine replacement therapy or combined bupropion and nicotine replacement therapy. MEASUREMENTS Survival analysis was used to model time to relapse using nicotine metabolism derived from CYP2A6 genotype-based estimates. Slow metabolism is defined as the lowest quartile of estimated metabolic function. FINDINGS CYP2A6-defined nicotine metabolic function moderated the effect of smoking cessation pharmacotherapy on smoking relapse over 90 days [hazard ratio (HR) = 2.81, 95% confidence interval (CI) = 1.32-5.99, P = 0.0075], with pharmacotherapy significantly slowing relapse in fast (HR = 0.39, 95% CI = 0.28-0.55, P = 1.97 × 10(-8)), but not slow metabolizers (HR = 1.09, 95% CI = 0.55-2.17, P = 0.80). Further, only the effect of nicotine replacement, and not bupropion, varies with CYP2A6-defined metabolic function. The effect of nicotine replacement on continuous abstinence is moderated by the combined genetic risks from CYP2A6 and CHRNA5 (Wald = 7.44, d.f. = 1, P = 0.0064). CONCLUSIONS Nicotine replacement therapy is effective among individuals with fast, but not slow, CYP2A6-defined nicotine metabolism. The effect of bupropion on relapse likelihood is unlikely to be affected by nicotine metabolism as estimated from CYP2A6 genotype. The variation in treatment responses among smokers with genes may guide future personalized smoking cessation interventions.
Collapse
Affiliation(s)
- Li-Shiun Chen
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - A. Joseph Bloom
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Timothy B. Baker
- Tobacco Research and Intervention, University of Wisconsin, School of Medicine, Madison, WI53711
| | - Stevens S. Smith
- Tobacco Research and Intervention, University of Wisconsin, School of Medicine, Madison, WI53711
| | - Megan E. Piper
- Tobacco Research and Intervention, University of Wisconsin, School of Medicine, Madison, WI53711
| | - Maribel Martinez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Nancy Saccone
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
| | - Dorothy Hatsukami
- Department of Psychiatry, University of Minnesota, Minneapolis, MN 55455
| | - Alison Goate
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Laura Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
29
|
Bloom AJ, Martinez M, Chen LS, Bierut LJ, Murphy SE, Goate A. CYP2B6 non-coding variation associated with smoking cessation is also associated with differences in allelic expression, splicing, and nicotine metabolism independent of common amino-acid changes. PLoS One 2013; 8:e79700. [PMID: 24260284 PMCID: PMC3829832 DOI: 10.1371/journal.pone.0079700] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 10/04/2013] [Indexed: 11/23/2022] Open
Abstract
The Cytochrome P450 2B6 (CYP2B6) enzyme makes a small contribution to hepatic nicotine metabolism relative to CYP2A6, but CYP2B6 is the primary enzyme responsible for metabolism of the smoking cessation drug bupropion. Using CYP2A6 genotype as a covariate, we find that a non-coding polymorphism in CYP2B6 previously associated with smoking cessation (rs8109525) is also significantly associated with nicotine metabolism. The association is independent of the well-studied non-synonymous variants rs3211371, rs3745274, and rs2279343 (CYP2B6*5 and *6). Expression studies demonstrate that rs8109525 is also associated with differences in CYP2B6 mRNA expression in liver biopsy samples. Splicing assays demonstrate that specific splice forms of CYP2B6 are associated with haplotypes defined by variants including rs3745274 and rs8109525. These results indicate differences in mRNA expression and splicing as potential molecular mechanisms by which non-coding variation in CYP2B6 may affect enzymatic activity leading to differences in metabolism and smoking cessation.
Collapse
Affiliation(s)
- A. Joseph Bloom
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| | - Maribel Martinez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Li-Shiun Chen
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Laura J. Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Sharon E. Murphy
- Department of Biochemistry Molecular Biology and BioPhysics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Alison Goate
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
30
|
Bloom AJ, Baker TB, Chen LS, Breslau N, Hatsukami D, Bierut LJ, Goate A. Variants in two adjacent genes, EGLN2 and CYP2A6, influence smoking behavior related to disease risk via different mechanisms. Hum Mol Genet 2013; 23:555-61. [PMID: 24045616 DOI: 10.1093/hmg/ddt432] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Genome-wide significant associations with cigarettes per day (CPD) and risk for lung cancer and chronic obstructive pulmonary disease (COPD) were previously reported in a region of 19q13, including CYP2A6 (nicotine metabolism enzyme) and EGLN2 (hypoxia response). The associated single nucleotide polymorphisms (SNPs) were assumed to be proxies for functional variation in CYP2A6. Here, we demonstrate that when CYP2A6 and EGLN2 genotypes are analyzed together, the key EGLN2 variant, rs3733829, is not associated with nicotine metabolism independent of CYP2A6, but is nevertheless independently associated with CPD, and with breath carbon monoxide (CO), a phenotype associated with cigarette consumption and relevant to hypoxia. SNPs in EGLN2 are also associated with nicotine dependence and with smoking efficiency (CO/CPD). These results indicate a previously unappreciated novel mechanism behind genome-wide significant associations with cigarette consumption and disease risk unrelated to nicotine metabolism.
Collapse
Affiliation(s)
- A Joseph Bloom
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
A compensatory effect upon splicing results in normal function of the CYP2A6*14 allele. Pharmacogenet Genomics 2013; 23:107-16. [PMID: 23292114 DOI: 10.1097/fpc.0b013e32835caf7d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A synonymous variant in the first exon of CYP2A6, rs1137115 (51G>A), defines the common reference allele CYP2A6*1A, and is associated with lower mRNA expression and slower in-vivo nicotine metabolism. Another common allele, CYP2A6*14, differs from CYP2A6*1A by a single variant, rs28399435 (86G>A, S29N). However, CYP2A6*14 shows in-vivo activity comparable with that of full-function alleles, and significantly higher than CYP2A6*1A. rs1137115A is predicted to create an exonic splicing suppressor site overlapping an exonic splicing enhancer (ESE) site in the first exon of CYP2A6, whereas rs28399435A is predicted to strengthen another adjacent ESE, potentially compensating for rs1137115A. Using an allelic expression assay to assess cDNAs produced from rs1137115 heterozygous liver biopsy samples, lower expression of the CYP2A6*1A allele is confirmed while CYP2A6*14 expression is found to be indistinguishable from that of rs1137115G alleles. Quantitative PCR assays to determine the relative abundance of spliced and unspliced or partially spliced CYP2A6 mRNAs in liver biopsy samples show that *1A/*1A homozygotes have a significantly lower ratio, due to both a reduction in spliced forms and an increase in unspliced or partially spliced CYP2A6. These results show the importance of common genetic variants that effect exonic splicing suppressor and ESEs to explain human variation regarding clinically-relevant phenotypes.
Collapse
|
32
|
Effects upon in-vivo nicotine metabolism reveal functional variation in FMO3 associated with cigarette consumption. Pharmacogenet Genomics 2013; 23:62-8. [PMID: 23211429 DOI: 10.1097/fpc.0b013e32835c3b48] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Flavin-containing monooxygenases (FMO) catalyze the metabolism of nucleophilic heteroatom-containing drugs and xenobiotics, including nicotine. Rare mutations in FMO3 are responsible for defective N-oxidation of dietary trimethylamine leading to trimethylaminuria, and common genetic variation in FMO3 has been linked to interindividual variability in metabolic function that may be substrate specific. METHODS A genetic model of CYP2A6 function is used as a covariate to reveal functional polymorphism in FMO3 that indirectly influences the ratio of deuterated nicotine metabolized to cotinine following oral administration. The association is tested between FMO3 haplotype and cigarette consumption in a set of nicotine-dependent smokers. RESULTS FMO3 haplotype, based on all common coding variants in Europeans, significantly predicts nicotine metabolism and accounts for ∼2% of variance in the apparent percent of nicotine metabolized to cotinine. The metabolic ratio is not associated with FMO2 haplotype or an FMO1 expression quantitative trait locus. Cross-validation demonstrates calculated FMO3 haplotype parameters to be robust and significantly improve the predictive nicotine metabolism model over CYP2A6 genotype alone. Functional classes of FMO3 haplotypes, as determined by their influence on nicotine metabolism to cotinine, are also significantly associated with cigarettes per day in nicotine-dependent European Americans (n=1025, P=0.04), and significantly interact (P=0.016) with CYP2A6 genotype to predict cigarettes per day. CONCLUSION These findings suggest that common polymorphisms in FMO3 influence nicotine clearance and that these genetic variants in turn influence cigarette consumption.
Collapse
|