1
|
Burke C, Taylor G, Freeman TP, Sallis H, Wootton RE, Munafò MR, Dardani C, Khouja J. Disentangling the effects of nicotine versus non-nicotine constituents of tobacco smoke on major depressive disorder: A multivariable Mendelian randomisation study. Addiction 2025; 120:1240-1252. [PMID: 39931798 PMCID: PMC12046462 DOI: 10.1111/add.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 01/15/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND AND AIMS There is growing evidence that tobacco smoking causes depression, but it is unclear which constituents of tobacco smoke (e.g. nicotine, carbon monoxide) may be responsible. We used Mendelian randomisation (MR) to measure the independent effect of nicotine on depression, by adjusting the effect of circulating nicotine exposure [via nicotine metabolite ratio (NMR)] for the overall effect of smoking heaviness [via cigarettes per day (CPD)] to account for the non-nicotine constituents of tobacco smoke. DESIGN Univariable MR and multivariable MR (MVMR) were used to measure the total and independent effects of genetic liability to NMR and CPD on major depressive disorder (MDD). Our primary method was inverse variance weighted (IVW) regression, with other methods as sensitivity analyses. SETTING AND PARTICIPANTS For the exposures, we used genome-wide association study (GWAS) summary statistics among European ancestry individuals for CPD (n = 143 210) and NMR (n = 5185). For the outcome, a GWAS of MDD stratified by smoking status was conducted using individual-level data from UK Biobank (n = 35 871-194 881). MEASUREMENTS Genetic variants associated with NMR (n = 6) and CPD (n = 53). FINDINGS Univariable MR-IVW indicated a causal effect of CPD on MDD [odds ratio (OR) = 1.13, 95% confidence interval (CI) = 1.04-1.23, P = 0.003] but no clear evidence for an effect of NMR on MDD (OR = 0.98, 95% CI = 0.97-1.00, P = 0.134). MVMR indicated a causal effect of CPD on MDD when accounting for NMR (IVW: OR = 1.19, 95% CI = 1.03-1.37, P = 0.017; Egger: OR = 1.13, 95% CI = 0.89-1.43, P = 0.300) and weak evidence of a small effect of NMR on MDD when accounting for CPD (IVW: OR = 0.98, 95% CI = 0.96-1.00, P = 0.057; Egger: OR = 0.98, 95% CI = 0.96-1.00, P = 0.038). CONCLUSIONS The role of nicotine exposure in risk of depression cannot be entirely dismissed. However, the causal effect of tobacco smoking increasing depression risk appears to be largely independent of circulating nicotine exposure, which implies the role of alternative causal pathways.
Collapse
Affiliation(s)
- Chloe Burke
- School of Psychological ScienceUniversity of BristolBristolUK
- Medical Research Council Integrative Epidemiology UnitUniversity of BristolBristolUK
- Department of PsychologyUniversity of BathBathUK
| | - Gemma Taylor
- Department of PsychologyUniversity of BathBathUK
| | | | - Hannah Sallis
- Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
| | - Robyn E. Wootton
- School of Psychological ScienceUniversity of BristolBristolUK
- Medical Research Council Integrative Epidemiology UnitUniversity of BristolBristolUK
- Lovisenberg Diakonale SykehusNic Waals InstituteOsloNorway
- PsychGen Centre for Genetic Epidemiology and Mental HealthNorwegian Institute of Public HealthOsloNorway
| | - Marcus R. Munafò
- School of Psychological ScienceUniversity of BristolBristolUK
- Medical Research Council Integrative Epidemiology UnitUniversity of BristolBristolUK
- NIHR Bristol Biomedical Research CentreBristolUK
| | - Christina Dardani
- Medical Research Council Integrative Epidemiology UnitUniversity of BristolBristolUK
| | - Jasmine Khouja
- School of Psychological ScienceUniversity of BristolBristolUK
- Medical Research Council Integrative Epidemiology UnitUniversity of BristolBristolUK
| |
Collapse
|
2
|
McKenzie C, Sloot B, Amorim FE, Robbins TW, Milton AL. Agonism at mGluR2 receptors reduces dysfunctional checking on a rodent analogue of compulsive-like checking in obsessive compulsive disorder. Psychopharmacology (Berl) 2025:10.1007/s00213-025-06774-2. [PMID: 40178609 DOI: 10.1007/s00213-025-06774-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 03/18/2025] [Indexed: 04/05/2025]
Abstract
RATIONALE Obsessive-compulsive disorder (OCD) affects 1-3% of the population. Current therapies, including selective serotonin reuptake inhibitors, are not universally effective in managing OCD. Recent discoveries indicating hyperactivation of key regions within the corticostriatal thalamic circuitry that supports OCD, and alterations in the ratio of glutamate: GABA in regions such as the anterior cingulate cortex, suggest that drugs targeting glutamatergic signalling may be effective in reducing OCD symptoms. OBJECTIVES This study sought to determine whether two drugs targeting metabotropic glutamate receptors could reduce excessive checking behaviour in a rodent analogue of compulsive-like checking in OCD, the Observing Response Task (ORT). METHODS Rats were trained on the ORT and separately classified on a pavlovian autoshaping task to identify the subpopulation of sign-trackers, which show higher levels of excessive checking. Once responding had stabilised, rats received systemic administration of different doses of the mGluR2 positive allosteric modulator AZD-8529 and its vehicle in a Latin square design, and the effects on ORT performance were assessed. Following completion of AZD-8529 dosing, a subset of rats received administration of different doses of the mGluR2/3 agonist LY404039 and its vehicle in a Latin square design, and ORT performance assessed. RESULTS Both AZD-8529 and LY404039 produced dose-dependent reductions in checking behaviour, including at doses that did not impair generalised measures of task performance. CONCLUSIONS The similarity in effect of AZD-8529 and LY404039 suggests that the capacity of these drugs to reduce checking is mediated by mGluR2s, which may provide a promising target for future treatment development for OCD.
Collapse
Affiliation(s)
- Colin McKenzie
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, CB2 3EB, UK
| | - Bart Sloot
- University of Radboud, Nijmegen, The Netherlands
| | | | - Trevor W Robbins
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, CB2 3EB, UK
| | - Amy L Milton
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, CB2 3EB, UK.
| |
Collapse
|
3
|
Cai J, Wang J, Wang Z, Wang J, Jia Y, Ma X. Perspectives on the α5 nicotinic acetylcholine receptor in lung cancer progression. Front Cell Dev Biol 2025; 13:1489958. [PMID: 40143965 PMCID: PMC11937065 DOI: 10.3389/fcell.2025.1489958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Nicotinic acetylcholine receptors (nAChRs) are widely expressed in a variety of cell types and are involved in multiple physiological regulatory mechanisms in cells, tissues and systems. Increasing evidence suggests that the α5 nicotinic acetylcholine receptor (α5-nAChR), encoded by the CHRNA5 gene, is one of a key mediator involved in lung cancer development and immune responses. Several studies have shown that it is a regulator that stimulates processes via various signaling pathways, including STAT3 in lung cancer. In addition, α5-nAChR has a profound effect on lung immune response through multiple immune-related factor pathways. In this review, we focus on the perspectives on α5-nAChR in lung cancer progression, which indicates that targeting α5-nAChR could provide novel anticancer and immune therapy strategies for lung cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaoli Ma
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
4
|
Woolf B, Cronjé HT, Zagkos L, Larsson SC, Gill D, Burgess S. Comparison of caffeine consumption behavior with plasma caffeine levels as exposure measures in drug-target mendelian randomization. Am J Epidemiol 2024; 193:1776-1784. [PMID: 38904434 PMCID: PMC7616520 DOI: 10.1093/aje/kwae143] [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: 05/30/2023] [Revised: 05/09/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024] Open
Abstract
Mendelian randomization is an epidemiologic technique that can explore the potential effect of perturbing a pharmacological target. Plasma caffeine levels can be used as a biomarker to measure the pharmacological effects of caffeine. Alternatively, this can be assessed using a behavioral proxy, such as average number of caffeinated drinks consumed per day. Either variable can be used as the exposure in a Mendelian randomization investigation, and to select which genetic variants to use as instrumental variables. Another possibility is to choose variants in gene regions with known biological relevance to caffeine level regulation. These choices affect the causal question that is being addressed by the analysis, and the validity of the analysis assumptions. Further, even when using the same genetic variants, the sign of Mendelian randomization estimates (positive or negative) can change depending on the choice of exposure. Some genetic variants that decrease caffeine metabolism associate with higher levels of plasma caffeine, but lower levels of caffeine consumption, as individuals with these variants require less caffeine consumption for the same physiological effect. We explore Mendelian randomization estimates for the effect of caffeine on body mass index, and discuss implications for variant and exposure choice in drug target Mendelian randomization investigations.
Collapse
Affiliation(s)
- Benjamin Woolf
- School of Psychological Science, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- MRC Biostatistics Unit at the University of Cambridge, Cambridge, UK
| | - Héléne T Cronjé
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark
| | - Loukas Zagkos
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2BX, United Kingdom
| | - Susanna C Larsson
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine Karolinska Institutet, Stockholm, Sweden
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London SW7 2BX, United Kingdom
| | - Stephen Burgess
- MRC Biostatistics Unit at the University of Cambridge, Cambridge, UK
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
5
|
Tochon L, Henkous N, Besson M, Maskos U, David V. Distinct Chrna5 mutations link excessive alcohol use to types I/II vulnerability profiles and IPN GABAergic neurons. Transl Psychiatry 2024; 14:461. [PMID: 39505853 PMCID: PMC11541707 DOI: 10.1038/s41398-024-03164-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 11/08/2024] Open
Abstract
Genome wide association and animal studies have implicated genetic variations in CHRNΑ5, encoding the α5 subunit-containing nicotinic acetylcholine receptors (α5*nAChRs), as a risk factor for developing alcohol use disorders (AUDs). To understand how α5*nAChR mutations may influence alcohol (EtOH) drinking behavior, we used a two-bottle choice procedure with intermittent access to alcohol in male and female transgenic mice expressing either the highly frequent human single nucleotide polymorphism (α5SNP/rs16969968) or a deletion of the Chrna5 gene (α5KO). AUDs-related preconsommatory traits (anxiety, sensation-seeking and impulsivity) were assessed with a battery of relevant tasks (elevated-plus maze, novel place preference and step-down inhibitory avoidance). The implication of the α5-expressing IPN GABAergic neurons in AUDs and related behavioral traits was verified using neurospecific lentiviral (LV)-induced reexpression of the α5 subunit in α5KOxGAD-Cre mice. Both α5SNP and α5KO mice showed over-consumption of EtOH, but displayed opposite vulnerability profiles consistent with Cloninger's subtypes of human AUDs. α5SNP mice showed Type I-like characteristics, i.e., high anxiety, novelty avoidance, whereas α5KOs exhibited Type II-like features such as low anxiety and high impulsivity. LV re-expression of the α5 subunit in IPN GABAergic neurons restored the control of EtOH intake and improved the impulsive phenotype. We demonstrate that the SNP (rs16969968) or null mutation of Chrna5 result in increased volitional EtOH consumption but opposite effects on anxiety, novelty-seeking and impulsive-like behaviors that match Cloninger type I and II of AUDs, including sex-related variations. IPN GABAergic neurons expressing α5*nAChRs play a key role in limiting both EtOH drinking and motor impulsivity.
Collapse
Affiliation(s)
- Léa Tochon
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, Bordeaux, France.
| | - Nadia Henkous
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, Bordeaux, France
| | - Morgane Besson
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Integrative Neurobiology of Cholinergic Systems, Paris, France
| | - Uwe Maskos
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Integrative Neurobiology of Cholinergic Systems, Paris, France
| | - Vincent David
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, Bordeaux, France.
| |
Collapse
|
6
|
Kang G, Song H, Bo L, Liu Q, Li Q, Li J, Pan P, Wang J, Jia Y, Sun H, Ma X. Nicotine promotes M2 macrophage polarization through α5-nAChR/SOX2/CSF-1 axis in lung adenocarcinoma. Cancer Immunol Immunother 2024; 74:11. [PMID: 39487876 PMCID: PMC11531455 DOI: 10.1007/s00262-024-03866-4] [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: 07/19/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024]
Abstract
α5-nicotinic acetylcholine receptor (α5-nAChR) plays a vital part in lung adenocarcinoma (LUAD). However, it is not comprehensively understood that how the α5-nAChR affects LUAD. Through diverse bioinformatics analyses and immunohistochemistry, the expressions of α5-nAChR and SOX2 as well as their relations were dissected. α5-nAChR regulated the differentiation of monocytes into M2 macrophages by targeting the STAT3/SOX2/CSF-1 signaling in the coculture system by western blotting and ChIP. α5-nAChR-mediated macrophage-mediated LUAD cell migration via SOX2/CSF-1 signaling in the cocultured medium. Correlations of α5-nAChR, SOX2 and M2 phenotype tumor-associated macrophages (TAMs) were validated in mouse LUAD models and clinical samples. α5-nAChR expression was connected to SOX2 expression, smoking and bad prognosis of LUAD among clinical samples. Nicotine-induced SOX2 expression was mediated by α5-nAChR via STAT3. Additionally, SOX2-mediated macrophage colony-stimulating factor (CSF-1) expression contributed to LUAD progression in vitro. Furthermore, α5-nAChR expression was strongly linked to pSTAT3, SOX2 and M2 macrophage marker CD206 expression and negatively correlated with M1 macrophage marker CD86 expression in vivo. It is indicated that M2 macrophages are mediated by the new α5-nAChR /SOX2/CSF-1 axis in nicotine-related LUAD, which is a potential therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Guiyu Kang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jiefang Road 105, Jinan, China
- Department of Medical Laboratory, Weifang Medical University, Weifang, China
- Department of Clinical Laboratory, The 960 Hospital of PLA, Jinan, China
| | - Hui Song
- Department of Cardiology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lei Bo
- Department of Clinical Laboratory, The 960 Hospital of PLA, Jinan, China
| | - Qi Liu
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jiefang Road 105, Jinan, China
| | - Qiang Li
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jiefang Road 105, Jinan, China
| | - Jingtan Li
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jiefang Road 105, Jinan, China
| | - Pan Pan
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jiefang Road 105, Jinan, China
| | - Jingting Wang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jiefang Road 105, Jinan, China
| | - Yanfei Jia
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jiefang Road 105, Jinan, China
| | - Haiji Sun
- College of Life Science, Shandong Normal University, Jinan, China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jiefang Road 105, Jinan, China.
- Department of Medical Laboratory, Weifang Medical University, Weifang, China.
| |
Collapse
|
7
|
Dixon P, Sallis H, Munafò M, Davey Smith G, Howe L. Mendelian Randomization Analysis of the Causal Effect of Cigarette Smoking on Hospital Costs. Nicotine Tob Res 2024; 26:1521-1529. [PMID: 38628153 PMCID: PMC11494471 DOI: 10.1093/ntr/ntae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/27/2024] [Accepted: 04/12/2024] [Indexed: 10/23/2024]
Abstract
INTRODUCTION Knowledge of the impact of smoking on health care costs is important for establishing the external effects of smoking and for evaluating policies intended to modify this behavior. Conventional analysis of this association is difficult because of omitted variable bias, reverse causality, and measurement error. AIMS AND METHODS We approached these challenges using a Mendelian Randomization study design; genetic variants associated with smoking behaviors were used in instrumental variables models with inpatient hospital costs (calculated from electronic health records) as the outcome. We undertook genome-wide association studies to identify genetic variants associated with smoking initiation and a composite smoking index (reflecting cumulative health impacts of smoking) on up to 300 045 individuals (mean age: 57 years at baseline, range 39-72 years) in the UK Biobank. We followed individuals up for a mean of 6 years. RESULTS Genetic liability to initiate smoking (ever vs. never smoking) was estimated to increase mean per-patient annual inpatient hospital costs by £477 (95% confidence interval (CI): £187 to £766). A one-unit change in genetic liability to the composite smoking index (range: 0-4.0) increased inpatient hospital costs by £204 (95% CI: £105 to £303) per unit increase in this index. There was some evidence that the composite smoking index causal models violated the instrumental variable assumptions, and all Mendelian Randomization models were estimated with considerable uncertainty. Models conditioning on risk tolerance were not robust to weak instrument bias. CONCLUSIONS Our findings have implications for the potential cost-effectiveness of smoking interventions. IMPLICATIONS We report the first Mendelian Randomization analysis of the causal effect of smoking on health care costs. Using two smoking phenotypes, we identified substantial impacts of smoking on inpatient hospital costs, although the causal models were associated with considerable uncertainty. These results could be used alongside other evidence on the impact of smoking to evaluate the cost-effectiveness of antismoking interventions and to understand the scale of externalities associated with this behavior.
Collapse
Affiliation(s)
- Padraig Dixon
- Nuffield Department of Primary Care Health Sciences, University of Oxford, UK
- MRC Integrative Epidemiology Unit, University of Bristol, UK
| | - Hannah Sallis
- MRC Integrative Epidemiology Unit, University of Bristol, UK
- School of Psychological Science, University of Bristol, UK
| | - Marcus Munafò
- MRC Integrative Epidemiology Unit, University of Bristol, UK
- School of Psychological Science, University of Bristol, UK
- NIHR Biomedical Research Centre, University of Bristol, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, UK
- NIHR Biomedical Research Centre, University of Bristol, UK
- Population Health Sciences, University of Bristol, UK
| | - Laura Howe
- MRC Integrative Epidemiology Unit, University of Bristol, UK
- Population Health Sciences, University of Bristol, UK
| |
Collapse
|
8
|
Patel A, DiTraglia FJ, Zuber V, Burgess S. Selecting Invalid Instruments to Improve Mendelian Randomization with Two-Sample Summary Data. Ann Appl Stat 2024; 18:23-aoas1856. [PMID: 38737575 PMCID: PMC7615940 DOI: 10.1214/23-aoas1856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Mendelian randomization (MR) is a widely-used method to estimate the causal relationship between a risk factor and disease. A fundamental part of any MR analysis is to choose appropriate genetic variants as instrumental variables. Genome-wide association studies often reveal that hundreds of genetic variants may be robustly associated with a risk factor, but in some situations investigators may have greater confidence in the instrument validity of only a smaller subset of variants. Nevertheless, the use of additional instruments may be optimal from the perspective of mean squared error even if they are slightly invalid; a small bias in estimation may be a price worth paying for a larger reduction in variance. For this purpose, we consider a method for "focused" instrument selection whereby genetic variants are selected to minimise the estimated asymptotic mean squared error of causal effect estimates. In a setting of many weak and locally invalid instruments, we propose a novel strategy to construct confidence intervals for post-selection focused estimators that guards against the worst case loss in asymptotic coverage. In empirical applications to: (i) validate lipid drug targets; and (ii) investigate vitamin D effects on a wide range of outcomes, our findings suggest that the optimal selection of instruments does not involve only a small number of biologically-justified instruments, but also many potentially invalid instruments.
Collapse
Affiliation(s)
| | | | - Verena Zuber
- Department of Biostatistics and Epidemiology, Imperial College London
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge
- Cardiovascular Epidemiology Unit, University of Cambridge
| |
Collapse
|
9
|
Jiang Y, Li H, Li Z, Du S, Zhang R, Zhao Y, Christiani DC, Shen S, Chen F. A cross-trait study of lung cancer and its related respiratory diseases based on large-scale exome sequencing population. Transl Lung Cancer Res 2024; 13:512-525. [PMID: 38601445 PMCID: PMC11002514 DOI: 10.21037/tlcr-24-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 02/27/2024] [Indexed: 04/12/2024]
Abstract
Background Genome-wide association studies (GWASs) explain the genetic susceptibility between diseases and common variants. Nevertheless, with the appearance of large-scale sequencing profiles, we could explore the rare coding variants in disease pathogenesis. Methods We estimated the genetic correlation of nine respiratory diseases and lung cancer in the UK Biobank (UKB) by linkage disequilibrium score regression (LDSC). Then, we performed exome-wide association studies at single-variant level and gene-level for lung cancer and lung cancer-related respiratory diseases using the whole-exome sequencing (WES) data of 427,934 European participants. Cross-trait meta-analysis was conducted by association analysis based on subsets (ASSET) to identify the pleiotropic variants, while in-silico functional analysis was performed to explore their function. Causal mediation analysis was used to explore whether these pleiotropic variants lead to lung cancer is mediated by affecting the chronic respiratory diseases. Results Five respiratory diseases [emphysema, pneumonia, asthma, chronic obstructive pulmonary disease (COPD), and fibrosis] were genetically correlated with lung cancer. We identified 102 significant independent variants at single-variant levels for lung cancer and five lung cancer-related diseases. 15:78590583:G>A (missense variant in CHRNA5) was shared in lung cancer, emphysema, and COPD. Meanwhile, 14 significant genes and 87 suggestive genes were identified in gene-based association tests, including HSD3B7 (lung cancer), SRSF2 (pneumonia), TNXB (asthma), TERT (fibrosis), MOSPD3 (emphysema). Based on the cross-trait meta-analysis, we detected 145 independent pleiotropic variants. We further identified abundant pathways with significant enrichment effects, demonstrating that these pleiotropic genes were functional. Meanwhile, the proportion of mediation effects of these variants ranged from 6 to 23 (emphysema: 23%; COPD: 20%; pneumonia: 20%; fibrosis: 7%; asthma: 6%) through these five respiratory diseases to the incidence of lung cancer. Conclusions The identified shared genetic variants, genes, biological pathways, and potential intermediate causal pathways provide a basis for further exploration of the relationship between lung cancer and respiratory diseases.
Collapse
Affiliation(s)
- Yunke Jiang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Hongru Li
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zaiming Li
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Sha Du
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ruyang Zhang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- China International Cooperation Center of Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Yang Zhao
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- China International Cooperation Center of Environment and Human Health, Nanjing Medical University, Nanjing, China
- Key Laboratory of Biomedical Big Data of Nanjing Medical University, Nanjing, China
| | - David C. Christiani
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Pulmonary and Critical Care Division, Massachusetts General Hospital, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Sipeng Shen
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- Key Laboratory of Biomedical Big Data of Nanjing Medical University, Nanjing, China
| | - Feng Chen
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- China International Cooperation Center of Environment and Human Health, Nanjing Medical University, Nanjing, China
- Key Laboratory of Biomedical Big Data of Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Kang G, Jiao Y, Pan P, Fan H, Li Q, Li X, Li J, Wang Y, Jia Y, Wang J, Sun H, Ma X. α5-nAChR/STAT3/CD47 axis contributed to nicotine-related lung adenocarcinoma progression and immune escape. Carcinogenesis 2023; 44:773-784. [PMID: 37681453 DOI: 10.1093/carcin/bgad061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/23/2023] [Accepted: 09/06/2023] [Indexed: 09/09/2023] Open
Abstract
OBJECTIVES The CHRNΑ5 gene, which encodes the α5-nicotinic acetylcholine receptor (α5-nAChR), is related to lung cancer and nicotine addiction. Smoking is closely related to the immunosuppressive effect of macrophages. CD47, a phagocytosis checkpoint in macrophages, is a therapeutic target in various cancer types. Nevertheless, the relationship between α5-nAChR and CD47 in lung cancer is still unclear. METHODS AND RESULTS The present study showed that α5-nAChR-mediated CD47 expression via STAT3 signaling, consequently leading to tumor progression and immune suppression in lung adenocarcinoma (LUAD). α5-nAChR expression was correlated with STAT3 expression, CD47 expression, smoking status and poor prognosis of LUAD in vivo. In vitro, α5-nAChR expression mediated the phosphorylation of STAT3, and phosphorylated STAT3 bound to the CD47 promoter and mediated CD47 expression. Downregulation of α5-nAChR and/or CD47 significantly reduced cell proliferation, migration, invasion, stemness and IL-10 expression, but increased TNF-α expression and phagocytosis of macrophages in LUAD. Furthermore, α5-nAChR/CD47 signaling contributed to the growth of subcutaneous xenograft tumors and liver metastasis of tumors in mice. CONCLUSION The α5-nAChR/STAT3/CD47 axis contributed to the progression and immune escape of lung cancer and may be a potential target for LUAD immunotherapy.
Collapse
Affiliation(s)
- Guiyu Kang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
- Department of Medical Laboratory, Weifang Medical University, Weifang, Shandong 261053, China
| | - Yang Jiao
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, China
| | - Pan Pan
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Huiping Fan
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Qiang Li
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Xiangying Li
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, China
| | - Jingtan Li
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, China
| | - Yan Wang
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, China
| | - Yanfei Jia
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Jingting Wang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
| | - Haiji Sun
- College of Life Science, Shandong Normal University, Jinan, Shandong 250014, China
| | - Xiaoli Ma
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, China
- Department of Medical Laboratory, Weifang Medical University, Weifang, Shandong 261053, China
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong University, Jinan, Shandong 250013, China
- Laboratory of Traditional Chinese Medicine & Stress Injury of Shandong Province, Jinan, Shandong 250013, China
| |
Collapse
|
11
|
Weiler M, Stieger KC, Shroff K, Klein JP, Wood WH, Zhang Y, Chandrasekaran P, Lehrmann E, Camandola S, Long JM, Mattson MP, Becker KG, Rapp PR. Transcriptional changes in the rat brain induced by repetitive transcranial magnetic stimulation. Front Hum Neurosci 2023; 17:1215291. [PMID: 38021223 PMCID: PMC10679736 DOI: 10.3389/fnhum.2023.1215291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 10/03/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Transcranial Magnetic Stimulation (TMS) is a noninvasive technique that uses pulsed magnetic fields to affect the physiology of the brain and central nervous system. Repetitive TMS (rTMS) has been used to study and treat several neurological conditions, but its complex molecular basis is largely unexplored. Methods Utilizing three experimental rat models (in vitro, ex vivo, and in vivo) and employing genome-wide microarray analysis, our study reveals the extensive impact of rTMS treatment on gene expression patterns. Results These effects are observed across various stimulation protocols, in diverse tissues, and are influenced by time and age. Notably, rTMS-induced alterations in gene expression span a wide range of biological pathways, such as glutamatergic, GABAergic, and anti-inflammatory pathways, ion channels, myelination, mitochondrial energetics, multiple neuron-and synapse-specific genes. Discussion This comprehensive transcriptional analysis induced by rTMS stimulation serves as a foundational characterization for subsequent experimental investigations and the exploration of potential clinical applications.
Collapse
Affiliation(s)
- Marina Weiler
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Kevin C. Stieger
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Kavisha Shroff
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Jessie P. Klein
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - William H. Wood
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Yongqing Zhang
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Prabha Chandrasekaran
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Elin Lehrmann
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Simonetta Camandola
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Jeffrey M. Long
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Kevin G. Becker
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Peter R. Rapp
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
12
|
Chau K, Raksadawan Y, Allison K, Ice JA, Scofield RH, Chepelev I, Harley ITW. Pervasive Sharing of Causal Genetic Risk Factors Contributes to Clinical and Molecular Overlap between Sjögren's Disease and Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:14449. [PMID: 37833897 PMCID: PMC10572278 DOI: 10.3390/ijms241914449] [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: 07/07/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 10/15/2023] Open
Abstract
SjD (Sjögren's Disease) and SLE (Systemic Lupus Erythematosus) are similar diseases. There is extensive overlap between the two in terms of both clinical features and pathobiologic mechanisms. Shared genetic risk is a potential explanation of this overlap. In this study, we evaluated whether these diseases share causal genetic risk factors. We compared the causal genetic risk for SLE and SjD using three complementary approaches. First, we examined the published GWAS results for these two diseases by analyzing the predicted causal gene protein-protein interaction networks of both diseases. Since this method does not account for overlapping risk intervals, we examined whether such intervals also overlap. Third, we used two-sample Mendelian randomization (two sample MR) using GWAS summary statistics to determine whether risk variants for SLE are causal for SjD and vice versa. We found that both the putative causal genes and the genomic risk intervals for SLE and SjD overlap 28- and 130-times more than expected by chance (p < 1.1 × 10-24 and p < 1.1 × 10-41, respectively). Further, two sample MR analysis confirmed that alone or in aggregate, SLE is likely causal for SjD and vice versa. [SjD variants predicting SLE: OR = 2.56; 95% CI (1.98-3.30); p < 1.4 × 10-13, inverse-variance weighted; SLE variants predicting SjD: OR = 1.36; 95% CI (1.26-1.47); p < 1.6 × 10-11, inverse-variance weighted]. Notably, some variants have disparate impact in terms of effect size across disease states. Overlapping causal genetic risk factors were found for both diseases using complementary approaches. These observations support the hypothesis that shared genetic factors drive the clinical and pathobiologic overlap between these diseases. Our study has implications for both differential diagnosis and future genetic studies of these two conditions.
Collapse
Affiliation(s)
- Karen Chau
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Yanint Raksadawan
- Internal Medicine Residency Program, Louis A. Weiss Memorial Hospital, Chicago, IL 60640, USA
| | - Kristen Allison
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - John A. Ice
- Research Service, Oklahoma City US Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - Robert Hal Scofield
- Research Service, Oklahoma City US Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
- Medicine Service, Oklahoma City US Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Iouri Chepelev
- Research Service, Cincinnati US Department of Veterans Affairs Medical Center, Cincinnati, OH 45220, USA
| | - Isaac T. W. Harley
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Rheumatology Section, Medicine Service, Eastern Colorado Healthcare System, US Department of Veterans Affairs Medical Center, Aurora, CO 80045, USA
| |
Collapse
|
13
|
Prom-Wormley EC, Wells JL, Landes L, Edmondson AN, Sankoh M, Jamieson B, Delk KJ, Surya S, Bhati S, Clifford J. A scoping review of smoking cessation pharmacogenetic studies to advance future research across racial, ethnic, and ancestral populations. Front Genet 2023; 14:1103966. [PMID: 37359362 PMCID: PMC10285878 DOI: 10.3389/fgene.2023.1103966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/25/2023] [Indexed: 06/28/2023] Open
Abstract
Abstinence rates among smokers attempting to quit remain low despite the wide availability and accessibility of pharmacological smoking cessation treatments. In addition, the prevalence of cessation attempts and abstinence differs by individual-level social factors such as race and ethnicity. Clinical treatment of nicotine dependence also continues to be challenged by individual-level variability in effectiveness to promote abstinence. The use of tailored smoking cessation strategies that incorporate information on individual-level social and genetic factors hold promise, although additional pharmacogenomic knowledge is still needed. In particular, genetic variants associated with pharmacological responses to smoking cessation treatment have generally been conducted in populations with participants that self-identify as White race or who are determined to be of European genetic ancestry. These results may not adequately capture the variability across all smokers as a result of understudied differences in allele frequencies across genetic ancestry populations. This suggests that much of the current pharmacogenetic study results for smoking cessation may not apply to all populations. Therefore, clinical application of pharmacogenetic results may exacerbate health inequities by racial and ethnic groups. This scoping review examines the extent to which racial, ethnic, and ancestral groups that experience differences in smoking rates and smoking cessation are represented in the existing body of published pharmacogenetic studies of smoking cessation. We will summarize results by race, ethnicity, and ancestry across pharmacological treatments and study designs. We will also explore current opportunities and challenges in conducting pharmacogenomic research on smoking cessation that encourages greater participant diversity, including practical barriers to clinical utilization of pharmacological smoking cessation treatment and clinical implementation of pharmacogenetic knowledge.
Collapse
Affiliation(s)
- Elizabeth C. Prom-Wormley
- Division of Epidemiology, Department of Family Medicine and Population Health, Virginia Commonwealth University, Richmond, VA, United States
| | - Jonathan L. Wells
- Division of Epidemiology, Department of Family Medicine and Population Health, Virginia Commonwealth University, Richmond, VA, United States
| | - Lori Landes
- Department of Family Medicine and Population Health, Virginia Commonwealth University, Richmond, VA, United States
| | - Amy N. Edmondson
- Division of Epidemiology, Department of Family Medicine and Population Health, Virginia Commonwealth University, Richmond, VA, United States
| | - Mariam Sankoh
- Department of Integrative Life Sciences, Virginia Commonwealth University, Richmond, VA, United States
| | - Brendan Jamieson
- Division of Epidemiology, Department of Family Medicine and Population Health, Virginia Commonwealth University, Richmond, VA, United States
| | - Kayla J. Delk
- Division of Epidemiology, Department of Family Medicine and Population Health, Virginia Commonwealth University, Richmond, VA, United States
| | - Sanya Surya
- Division of Epidemiology, Department of Family Medicine and Population Health, Virginia Commonwealth University, Richmond, VA, United States
| | - Shambhavi Bhati
- Division of Epidemiology, Department of Family Medicine and Population Health, Virginia Commonwealth University, Richmond, VA, United States
| | - James Clifford
- Department of Public Health, Brody School of Medicine, East Carolina University, Greenville, United States
| |
Collapse
|
14
|
Kamens HM, Flarend G, Horton WJ. The role of nicotinic receptors in alcohol consumption. Pharmacol Res 2023; 190:106705. [PMID: 36813094 PMCID: PMC10083870 DOI: 10.1016/j.phrs.2023.106705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/14/2023] [Accepted: 02/19/2023] [Indexed: 02/22/2023]
Abstract
The use of alcohol causes significant morbidity and mortality across the globe. Alcohol use disorder (AUD) is defined by the excessive use of this drug despite a negative impact on the individual's life. While there are currently medications available to treat AUD, they have limited efficacy and several side effects. As such, it is essential to continue to look for novel therapeutics. One target for novel therapeutics is nicotinic acetylcholine receptors (nAChRs). Here we systematically review the literature on the involvement of nAChRs in alcohol consumption. Data from both genetic and pharmacology studies provide evidence that nAChRs modulate alcohol intake. Interestingly, pharmacological modulation of all nAChR subtypes examined can decrease alcohol consumption. The reviewed literature demonstrates that nAChRs should continue to be investigated as novel therapeutics for AUD.
Collapse
Affiliation(s)
- Helen M Kamens
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA 16802, United States.
| | - Geneva Flarend
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA 16802, United States
| | - William J Horton
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA 16802, United States
| |
Collapse
|
15
|
Lor CS, Haugg A, Zhang M, Schneider L, Herdener M, Quednow BB, Golestani N, Scharnowski F. Thalamic volume and functional connectivity are associated with nicotine dependence severity and craving. Addict Biol 2023; 28:e13261. [PMID: 36577730 PMCID: PMC10078543 DOI: 10.1111/adb.13261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022]
Abstract
Tobacco smoking is associated with deleterious health outcomes. Most smokers want to quit smoking, yet relapse rates are high. Understanding neural differences associated with tobacco use may help generate novel treatment options. Several animal studies have recently highlighted the central role of the thalamus in substance use disorders, but this research focus has been understudied in human smokers. Here, we investigated associations between structural and functional magnetic resonance imaging measures of the thalamus and its subnuclei to distinct smoking characteristics. We acquired anatomical scans of 32 smokers as well as functional resting-state scans before and after a cue-reactivity task. Thalamic functional connectivity was associated with craving and dependence severity, whereas the volume of the thalamus was associated with dependence severity only. Craving, which fluctuates rapidly, was best characterized by differences in brain function, whereas the rather persistent syndrome of dependence severity was associated with both brain structural differences and function. Our study supports the notion that functional versus structural measures tend to be associated with behavioural measures that evolve at faster versus slower temporal scales, respectively. It confirms the importance of the thalamus to understand mechanisms of addiction and highlights it as a potential target for brain-based interventions to support smoking cessation, such as brain stimulation and neurofeedback.
Collapse
Affiliation(s)
- Cindy Sumaly Lor
- Department of Cognition, Emotion, and Methods in Psychology, University of Vienna, Vienna, Austria.,Department of Psychiatry, Psychotherapy and Psychosomatics
- Psychiatric University Hospital Zurich, University of Zurich, Zürich, Switzerland
| | - Amelie Haugg
- Department of Child and Adolescent Psychiatry and Psychotherapy
- Psychiatric University Hospital Zurich, University of Zurich, Zürich, Switzerland
| | - Mengfan Zhang
- Department of Cognition, Emotion, and Methods in Psychology, University of Vienna, Vienna, Austria.,Department of Psychiatry, Psychotherapy and Psychosomatics
- Psychiatric University Hospital Zurich, University of Zurich, Zürich, Switzerland
| | - Letitia Schneider
- Division of Psychology and Language Sciences, University College London, London, United Kingdom
| | - Marcus Herdener
- Department of Psychiatry, Psychotherapy and Psychosomatics
- Psychiatric University Hospital Zurich, University of Zurich, Zürich, Switzerland
| | - Boris B Quednow
- Department of Psychiatry, Psychotherapy and Psychosomatics
- Psychiatric University Hospital Zurich, University of Zurich, Zürich, Switzerland
| | - Narly Golestani
- Brain and Language Lab
- Cognitive Science Hub, University of Vienna, Vienna, Austria.,Department of Behavioral and Cognitive Biology, University of Vienna, Vienna, Austria.,Department of Psychology
- Faculty of Psychology and Educational Sciences, University of Geneva, Geneva, Switzerland
| | - Frank Scharnowski
- Department of Cognition, Emotion, and Methods in Psychology, University of Vienna, Vienna, Austria.,Department of Psychiatry, Psychotherapy and Psychosomatics
- Psychiatric University Hospital Zurich, University of Zurich, Zürich, Switzerland
| |
Collapse
|
16
|
Lahmar Z, Ahmed E, Fort A, Vachier I, Bourdin A, Bergougnoux A. Hedgehog pathway and its inhibitors in chronic obstructive pulmonary disease (COPD). Pharmacol Ther 2022; 240:108295. [PMID: 36191777 DOI: 10.1016/j.pharmthera.2022.108295] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/22/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022]
Abstract
COPD affects millions of people and is now ranked as the third leading cause of death worldwide. This largely untreatable chronic airway disease results in irreversible destruction of lung architecture. The small lung hypothesis is now supported by epidemiological, physiological and clinical studies. Accordingly, the early and severe COPD phenotype carries the most dreadful prognosis and finds its roots during lung growth. Pathophysiological mechanisms remain poorly understood and implicate individual susceptibility (genetics), a large part of environmental factors (viral infections, tobacco consumption, air pollution) and the combined effects of those triggers on gene expression. Genetic susceptibility is most likely involved as the disease is severe and starts early in life. The latter observation led to the identification of Mendelian inheritance via disease-causing variants of SERPINA1 - known as the basis for alpha-1 anti-trypsin deficiency, and TERT. In the last two decades multiple genome wide association studies (GWAS) identified many single nucleotide polymorphisms (SNPs) associated with COPD. High significance SNPs are located in 4q31 near HHIP which encodes an evolutionarily highly conserved physiological inhibitor of the Hedgehog signaling pathway (HH). HHIP is critical to several in utero developmental lung processes. It is also implicated in homeostasis, injury response, epithelial-mesenchymal transition and tumor resistance to apoptosis. A few studies have reported decreased HHIP RNA and protein levels in human adult COPD lungs. HHIP+/- murine models led to emphysema. HH pathway inhibitors, such as vismodegib and sonidegib, are already validated in oncology, whereas other drugs have evidenced in vitro effects. Targeting the Hedgehog pathway could lead to a new therapeutic avenue in COPD. In this review, we focused on the early and severe COPD phenotype and the small lung hypothesis by exploring genetic susceptibility traits that are potentially treatable, thus summarizing promising therapeutics for the future.
Collapse
Affiliation(s)
- Z Lahmar
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France
| | - E Ahmed
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France; PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - A Fort
- PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - I Vachier
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France; PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - A Bourdin
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France; PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - A Bergougnoux
- PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France; Laboratoire de Génétique Moléculaire et de Cytogénomique, CHU de Montpellier, Montpellier, France.
| |
Collapse
|
17
|
Neandertal introgression partitions the genetic landscape of neuropsychiatric disorders and associated behavioral phenotypes. Transl Psychiatry 2022; 12:433. [PMID: 36198681 PMCID: PMC9534885 DOI: 10.1038/s41398-022-02196-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/13/2022] [Accepted: 09/20/2022] [Indexed: 11/08/2022] Open
Abstract
Despite advances in identifying the genetic basis of psychiatric and neurological disorders, fundamental questions about their evolutionary origins remain elusive. Here, introgressed variants from archaic humans such as Neandertals can serve as an intriguing research paradigm. We compared the number of associations for Neandertal variants to the number of associations of frequency-matched non-archaic variants with regard to human CNS disorders (neurological and psychiatric), nervous system drug prescriptions (as a proxy for disease), and related, non-disease phenotypes in the UK biobank (UKBB). While no enrichment for Neandertal genetic variants were observed in the UKBB for psychiatric or neurological disease categories, we found significant associations with certain behavioral phenotypes including pain, chronotype/sleep, smoking and alcohol consumption. In some instances, the enrichment signal was driven by Neandertal variants that represented the strongest association genome-wide. SNPs within a Neandertal haplotype that was associated with smoking in the UKBB could be replicated in four independent genomics datasets.Our data suggest that evolutionary processes in recent human evolution like admixture with Neandertals significantly contribute to behavioral phenotypes but not psychiatric and neurological diseases. These findings help to link genetic variants in a population to putative past beneficial effects, which likely only indirectly contribute to pathology in modern day humans.
Collapse
|
18
|
Luessen DJ, Conn PJ. Allosteric Modulators of Metabotropic Glutamate Receptors as Novel Therapeutics for Neuropsychiatric Disease. Pharmacol Rev 2022; 74:630-661. [PMID: 35710132 PMCID: PMC9553119 DOI: 10.1124/pharmrev.121.000540] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors, a family of G-protein-coupled receptors, have been identified as novel therapeutic targets based on extensive research supporting their diverse contributions to cell signaling and physiology throughout the nervous system and important roles in regulating complex behaviors, such as cognition, reward, and movement. Thus, targeting mGlu receptors may be a promising strategy for the treatment of several brain disorders. Ongoing advances in the discovery of subtype-selective allosteric modulators for mGlu receptors has provided an unprecedented opportunity for highly specific modulation of signaling by individual mGlu receptor subtypes in the brain by targeting sites distinct from orthosteric or endogenous ligand binding sites on mGlu receptors. These pharmacological agents provide the unparalleled opportunity to selectively regulate neuronal excitability, synaptic transmission, and subsequent behavioral output pertinent to many brain disorders. Here, we review preclinical and clinical evidence supporting the utility of mGlu receptor allosteric modulators as novel therapeutic approaches to treat neuropsychiatric diseases, such as schizophrenia, substance use disorders, and stress-related disorders. SIGNIFICANCE STATEMENT: Allosteric modulation of metabotropic glutamate (mGlu) receptors represents a promising therapeutic strategy to normalize dysregulated cellular physiology associated with neuropsychiatric disease. This review summarizes preclinical and clinical studies using mGlu receptor allosteric modulators as experimental tools and potential therapeutic approaches for the treatment of neuropsychiatric diseases, including schizophrenia, stress, and substance use disorders.
Collapse
|
19
|
CHRNA5 rs16969968 and CHRNA3 rs578776 polymorphisms are associated with multiple nicotine dependence phenotypes in Bangladeshi smokers. Heliyon 2022; 8:e09947. [PMID: 35865987 PMCID: PMC9293740 DOI: 10.1016/j.heliyon.2022.e09947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/14/2021] [Accepted: 07/08/2022] [Indexed: 11/23/2022] Open
Abstract
Background A recent study has identified the role of CHRNA5-A3-B4 gene cluster variants rs16969968 and rs578776 of nicotinic acetylcholine receptors (nAChRs) on smoking status in Bengali ethnicity. The aim of the current study was to investigate whether these rs16969968-rs578776-rs11072768 single nucleotide polymorphisms (SNPs) of CHRNA5-A3-B4 gene cluster were associated with nicotine dependence (ND) and related phenotypes. Methods The Fagerstrom Test for Nicotine Dependence (FTND) and Cigarette Dependence Scale (CDS-12) were used to assess the degree of ND, and genotyping was done using the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method on a cohort of 129 male smokers participating in a structured questionnaire-based survey. Results Smokers with AA genotype of CHRNA5 rs16969968 SNP were at significantly increased risk of developing ND compared to its wild type variant with odds ratio (ORs) of 1.20 (FTND: 95% CI 0.25–5.37, p = 0.253) and 2.48 (CDS-12: 95% CI 0.46–13.26, p = 0.081), respectively. Conversely, smokers with AA genotype of CHRNA3 rs578776 variant had a strong protective effect against ND development (ORs = 0.27, 95% CI 0.09–0.80, p = 0.076). There was no such link reported in CHRNB4 rs11072768 variant carriers. Similarly, G-A/G-A diplotype of rs16969968_rs578776 variants was discovered to be a protective factor against ND. Moreover, demographic features such as age, occupation and dwelling status were found to be significantly associated with ND. Conclusion Taken together, CHRNA5-A3-B4 gene cluster variants rs16969968 and rs578776 as well as specific demographic characteristics regulate ND and related smoking phenotypes in Bangladeshi male smokers. Further studies with large sample sizes are required to substantially validate the significance.
Collapse
|
20
|
Breum AW, Falk S, Svendsen CSA, Nicolaisen TS, Mathiesen CV, Maskos U, Clemmensen C. Divergent Roles of α5 and β4 Nicotinic Receptor Subunits in Food Reward and Nicotine-induced Weight Loss in Male Mice. Endocrinology 2022; 163:6590007. [PMID: 35595472 PMCID: PMC9217964 DOI: 10.1210/endocr/bqac079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Indexed: 11/19/2022]
Abstract
A major obstacle to successful smoking cessation is the prospect of weight gain. Despite a clear relationship between cigarette smoking and body weight, surprisingly little is known about the physiological and molecular mechanism by which nicotine affects energy homeostasis and food-motivated behaviors. Here we use loss-of-function mouse models to demonstrate that 2 nicotinic acetylcholine receptor (nAChR) subunits encoded by the CHRNA5-CHRNA3-CHRNB4 gene cluster, α5 and β4, exhibit divergent roles in food reward. We also reveal that β4-containing nAChRs are essential for the weight-lowering effects of nicotine in diet-induced obese mice. Finally, our data support the notion of crosstalk between incretin biology and nAChR signaling, as we demonstrate that the glycemic benefits of glucagon-like peptide-1 receptor activation partially relies on β4-containing nAChRs. Together, these data encourage further research into the role of cholinergic neurotransmission in regulating food reward and the translational pursuit of site-directed targeting of β4-containing nAChRs for treatment of metabolic disease.
Collapse
Affiliation(s)
| | | | - Charlotte Sashi Aier Svendsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Trine Sand Nicolaisen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie Vad Mathiesen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Uwe Maskos
- Institut Pasteur, Université de Paris, Integrative Neurobiology of Cholinergic Systems, CNRS UMR 3571, Paris, France
| | - Christoffer Clemmensen
- Correspondence: Christoffer Clemmensen, Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
21
|
Abstract
Mendelian randomization (MR) is a method of studying the causal effects of modifiable exposures (i.e., potential risk factors) on health, social, and economic outcomes using genetic variants associated with the specific exposures of interest. MR provides a more robust understanding of the influence of these exposures on outcomes because germline genetic variants are randomly inherited from parents to offspring and, as a result, should not be related to potential confounding factors that influence exposure-outcome associations. The genetic variant can therefore be used as a tool to link the proposed risk factor and outcome, and to estimate this effect with less confounding and bias than conventional epidemiological approaches. We describe the scope of MR, highlighting the range of applications being made possible as genetic data sets and resources become larger and more freely available. We outline the MR approach in detail, covering concepts, assumptions, and estimation methods. We cover some common misconceptions, provide strategies for overcoming violation of assumptions, and discuss future prospects for extending the clinical applicability, methodological innovations, robustness, and generalizability of MR findings.
Collapse
Affiliation(s)
- Rebecca C Richmond
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, United Kingdom
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol NHS Foundation Trust and University of Bristol, Bristol BS1 3NU, United Kingdom
| |
Collapse
|
22
|
Ye Z, Mo C, Ke H, Yan Q, Chen C, Kochunov P, Hong LE, Mitchell BD, Chen S, Ma T. Meta-Analysis of Transcriptome-Wide Association Studies across 13 Brain Tissues Identified Novel Clusters of Genes Associated with Nicotine Addiction. Genes (Basel) 2021; 13:37. [PMID: 35052378 PMCID: PMC8775257 DOI: 10.3390/genes13010037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/14/2021] [Accepted: 12/18/2021] [Indexed: 12/01/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified and reproduced thousands of diseases associated loci, but many of them are not directly interpretable due to the strong linkage disequilibrium among variants. Transcriptome-wide association studies (TWAS) incorporated expression quantitative trait loci (eQTL) cohorts as a reference panel to detect associations with the phenotype at the gene level and have been gaining popularity in recent years. For nicotine addiction, several important susceptible genetic variants were identified by GWAS, but TWAS that detected genes associated with nicotine addiction and unveiled the underlying molecular mechanism were still lacking. In this study, we used eQTL data from the Genotype-Tissue Expression (GTEx) consortium as a reference panel to conduct tissue-specific TWAS on cigarettes per day (CPD) over thirteen brain tissues in two large cohorts: UK Biobank (UKBB; number of participants (N) = 142,202) and the GWAS & Sequencing Consortium of Alcohol and Nicotine use (GSCAN; N = 143,210), then meta-analyzing the results across tissues while considering the heterogeneity across tissues. We identified three major clusters of genes with different meta-patterns across tissues consistent in both cohorts, including homogenous genes associated with CPD in all brain tissues; partially homogeneous genes associated with CPD in cortex, cerebellum, and hippocampus tissues; and, lastly, the tissue-specific genes associated with CPD in only a few specific brain tissues. Downstream enrichment analyses on each gene cluster identified unique biological pathways associated with CPD and provided important biological insights into the regulatory mechanism of nicotine dependence in the brain.
Collapse
Affiliation(s)
- Zhenyao Ye
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (Z.Y.); (C.M.); (P.K.); (L.E.H.)
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD 21201, USA;
| | - Chen Mo
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (Z.Y.); (C.M.); (P.K.); (L.E.H.)
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD 21201, USA;
| | - Hongjie Ke
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20742, USA;
| | - Qi Yan
- Irving Medical Center, Department of Obstetrics & Gynecology, Columbia University, New York, NY 10032, USA;
| | - Chixiang Chen
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD 21201, USA;
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (Z.Y.); (C.M.); (P.K.); (L.E.H.)
| | - L. Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (Z.Y.); (C.M.); (P.K.); (L.E.H.)
| | - Braxton D. Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Shuo Chen
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (Z.Y.); (C.M.); (P.K.); (L.E.H.)
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD 21201, USA;
| | - Tianzhou Ma
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20742, USA;
| |
Collapse
|
23
|
Gelernter J, Polimanti R. Genetics of substance use disorders in the era of big data. Nat Rev Genet 2021; 22:712-729. [PMID: 34211176 PMCID: PMC9210391 DOI: 10.1038/s41576-021-00377-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2021] [Indexed: 02/06/2023]
Abstract
Substance use disorders (SUDs) are conditions in which the use of legal or illegal substances, such as nicotine, alcohol or opioids, results in clinical and functional impairment. SUDs and, more generally, substance use are genetically complex traits that are enormously costly on an individual and societal basis. The past few years have seen remarkable progress in our understanding of the genetics, and therefore the biology, of substance use and abuse. Various studies - including of well-defined phenotypes in deeply phenotyped samples, as well as broadly defined phenotypes in meta-analysis and biobank samples - have revealed multiple risk loci for these common traits. A key emerging insight from this work establishes a biological and genetic distinction between quantity and/or frequency measures of substance use (which may involve low levels of use without dependence), versus symptoms related to physical dependence.
Collapse
Affiliation(s)
- Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT, USA.
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA.
| | - Renato Polimanti
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT, USA
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare Center, West Haven, CT, USA
| |
Collapse
|
24
|
Ye Z, Mo C, Liu S, Hatch KS, Gao S, Ma Y, Hong LE, Thompson PM, Jahanshad N, Acheson A, Garavan H, Shen L, Nichols TE, Kochunov P, Chen S, Ma T. White Matter Integrity and Nicotine Dependence: Evaluating Vertical and Horizontal Pleiotropy. Front Neurosci 2021; 15:738037. [PMID: 34720862 PMCID: PMC8551454 DOI: 10.3389/fnins.2021.738037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/01/2021] [Indexed: 01/26/2023] Open
Abstract
Tobacco smoking is an addictive behavior that supports nicotine dependence and is an independent risk factor for cancer and other illnesses. Its neurogenetic mechanisms are not fully understood but may act through alterations in the cerebral white matter (WM). We hypothesized that the vertical pleiotropic pathways, where genetic variants influence a trait that in turn influences another trait, link genetic factors, integrity of cerebral WM, and nicotine addiction. We tested this hypothesis using individual genetic factors, WM integrity measured by fractional anisotropy (FA), and nicotine dependence-related smoking phenotypes, including smoking status (SS) and cigarettes per day (CPDs), in a large epidemiological sample collected by the UK Biobank. We performed a genome-wide association study (GWAS) to identify previously reported loci associated with smoking behavior. Smoking was found to be associated with reduced WM integrity in multiple brain regions. We then evaluated two competing vertical pathways: Genes → WM integrity → Smoking versus Genes → Smoking → WM integrity and a horizontal pleiotropy pathway where genetic factors independently affect both smoking and WM integrity. The causal pathway analysis identified 272 pleiotropic single-nucleotide polymorphisms (SNPs) whose effects on SS were mediated by FA, as well as 22 pleiotropic SNPs whose effects on FA were mediated by CPD. These SNPs were mainly located in important susceptibility genes for smoking-induced diseases NCAM1 and IREB2. Our findings revealed the role of cerebral WM in the maintenance of the complex addiction and provided potential genetic targets for future research in examining how changes in WM integrity contribute to the nicotine effects on the brain.
Collapse
Affiliation(s)
- Zhenyao Ye
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Chen Mo
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Song Liu
- School of Computer Science and Technology, Qilu University of Technology, Shandong Academy of Sciences, Jinan, China
| | - Kathryn S Hatch
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Si Gao
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Yizhou Ma
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - L Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| | - Ashley Acheson
- Department of Psychiatry and Behavioral Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Hugh Garavan
- Department of Psychiatry, The University of Vermont, Burlington, VT, United States
| | - Li Shen
- Department of Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Thomas E Nichols
- Oxford Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Shuo Chen
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| | - Tianzhou Ma
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, College Park, MD, United States
| |
Collapse
|
25
|
El-Boraie A, Tyndale RF. The Role of Pharmacogenetics in Smoking. Clin Pharmacol Ther 2021; 110:599-606. [PMID: 34165800 DOI: 10.1002/cpt.2345] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/21/2021] [Indexed: 11/09/2022]
Abstract
Smoking continues to be the leading preventable contributor to death worldwide. Twin studies have suggested a significant genetic contribution underlying most smoking behaviors (40-70% heritability estimates). Candidate gene studies of smoking phenotypes have identified several pharmacogenes implicated in nicotine's pharmacokinetics (CYP2A6, CYP2B6, CYP2A13, FMOs, UGTs, and OCT2), and nicotine's pharmacodynamic response in the central nervous system (nicotinic acetylcholine receptors, as well as through the dopaminergic and serotonergic systems). Subsequent genome-wide association studies (GWAS) have confirmed the role of certain pharmacogenes through hypothesis-free approaches. Furthermore, pharmacogenes that alter the efficacy of smoking cessation pharmacotherapies, including nicotine replacement therapies, bupropion, and varenicline, may also impact quitting success. In this brief review we highlight the role of pharmacogenes in smoking behaviors, such as smoking status, consumption, nicotine dependence, spontaneous quitting, and altered abstinence to pharmacotherapies; We provide examples from initial candidate gene associations and subsequent GWAS. The genes CYP2A6 and the CHRNA5-A3-B4 confer the most replicated sources of genetic variation in smoking behaviors, likely due to their importance in nicotine's pharmacology. We will also provide examples of genetic scoring approaches, and the role of rare variants in explaining a portion of the missing heritability in smoking behaviors.
Collapse
Affiliation(s)
- Ahmed El-Boraie
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Rachel F Tyndale
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.,Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Genetically Targeted Connectivity Tracing Excludes Dopaminergic Inputs to the Interpeduncular Nucleus from the Ventral Tegmentum and Substantia Nigra. eNeuro 2021; 8:ENEURO.0127-21.2021. [PMID: 34088738 PMCID: PMC8223495 DOI: 10.1523/eneuro.0127-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 12/15/2022] Open
Abstract
The “habenulopeduncular system” consists of the medial habenula (MHb) and its principal target of innervation, the interpeduncular nucleus (IP). Neurons in the ventral MHb (MHbV) express acetylcholine along with glutamate, and both the MHb and IP are rich in nicotinic acetylcholine receptors. Much of the work on this system has focused on nicotinic mechanisms and their clinical implications for nicotine use, particularly because the IP expresses the α5 nicotinic receptor subunit, encoded by the CHRNA5 gene, which is genetically linked to smoking risk. A working model has emerged in which nicotine use may be determined by the balance of reinforcement mediated in part by nicotine effects on dopamine reward pathways, and an aversive “brake” on nicotine consumption encoded in the MHb-IP pathway. However, recent work has proposed that the IP also receives direct dopaminergic input from the ventral tegmental area (VTA). If correct, this would significantly impact the prevailing model of IP function. Here, we have used Chrna5Cre mice to perform rabies virus-mediated retrograde tracing of global inputs to the IP. We have also used Cre-dependent adeno-associated virus (AAV) anterograde tracing using Slc6a3Cre (DATCre) mice to map VTA dopaminergic efferents, and we have examined tract-tracing data using other transgenic models for dopaminergic neurons available in a public database. Consistent with the existing literature using non-genetic tracing methods, none of these experiments show a significant anatomic connection from the VTA or substantia nigra (SN) to the IP, and thus do not support a model of direct dopaminergic input to the habenulopeduncular system.
Collapse
|
27
|
Yao C, Joehanes R, Wilson R, Tanaka T, Ferrucci L, Kretschmer A, Prokisch H, Schramm K, Gieger C, Peters A, Waldenberger M, Marzi C, Herder C, Levy D. Epigenome-wide association study of whole blood gene expression in Framingham Heart Study participants provides molecular insight into the potential role of CHRNA5 in cigarette smoking-related lung diseases. Clin Epigenetics 2021; 13:60. [PMID: 33752734 PMCID: PMC7986283 DOI: 10.1186/s13148-021-01041-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 02/28/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND DNA methylation is a key epigenetic modification that can directly affect gene regulation. DNA methylation is highly influenced by environmental factors such as cigarette smoking, which is causally related to chronic obstructive pulmonary disease (COPD) and lung cancer. To date, there have been few large-scale, combined analyses of DNA methylation and gene expression and their interrelations with lung diseases. RESULTS We performed an epigenome-wide association study of whole blood gene expression in ~ 6000 individuals from four cohorts. We discovered and replicated numerous CpGs associated with the expression of cis genes within 500 kb of each CpG, with 148 to 1,741 cis CpG-transcript pairs identified across cohorts. We found that the closer a CpG resided to a transcription start site, the larger its effect size, and that 36% of cis CpG-transcript pairs share the same causal genetic variant. Mendelian randomization analyses revealed that hypomethylation and lower expression of CHRNA5, which encodes a smoking-related nicotinic receptor, are causally linked to increased risk of COPD and lung cancer. This putatively causal relationship was further validated in lung tissue data. CONCLUSIONS Our results provide a large and comprehensive association study of whole blood DNA methylation with gene expression. Expression platform differences rather than population differences are critical to the replication of cis CpG-transcript pairs. The low reproducibility of trans CpG-transcript pairs suggests that DNA methylation regulates nearby rather than remote gene expression. The putatively causal roles of methylation and expression of CHRNA5 in relation to COPD and lung cancer provide evidence for a mechanistic link between patterns of smoking-related epigenetic variation and lung diseases, and highlight potential therapeutic targets for lung diseases and smoking cessation.
Collapse
Affiliation(s)
- Chen Yao
- The Framingham Heart Study, 73 Mt. Wayte Avenue, Framingham, MA, 01702, USA
- The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Roby Joehanes
- The Framingham Heart Study, 73 Mt. Wayte Avenue, Framingham, MA, 01702, USA
- The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Rory Wilson
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Bavaria, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Bavaria, Neuherberg, Germany
| | - Toshiko Tanaka
- Longitudinal Study Section, National Institute On Aging, Baltimore, MD, USA
| | - Luigi Ferrucci
- Longitudinal Study Section, National Institute On Aging, Baltimore, MD, USA
| | - Anja Kretschmer
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Bavaria, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Bavaria, Neuherberg, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technical University Munich, München, Germany
- Institute for Neurogenomics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Katharina Schramm
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich, 81377, Munich, Germany
- Department of Internal Medicine I (Cardiology), Hospital of the Ludwig-Maximilians-University (LMU) Munich, 81377, Munich, Germany
| | - Christian Gieger
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Bavaria, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Bavaria, Neuherberg, Germany
| | - Annette Peters
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Bavaria, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Bavaria, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Bavaria, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Bavaria, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Carola Marzi
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, German Research Center for Environmental Health, Neuherberg, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research At Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Daniel Levy
- The Framingham Heart Study, 73 Mt. Wayte Avenue, Framingham, MA, 01702, USA.
- The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA.
| |
Collapse
|
28
|
Weltzin MM, George AA, Lukas RJ, Whiteaker P. Sleep-related hypermotor epilepsy associated mutations uncover important kinetic roles of α4β2- nicotinic acetylcholine receptor intracellular structures. PLoS One 2021; 16:e0247825. [PMID: 33657187 PMCID: PMC7928491 DOI: 10.1371/journal.pone.0247825] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
Sleep-related hypermotor epilepsy (SHE) is a group of seizure disorders prominently associated with mutations in nicotinic acetylcholine receptors (nAChR). The most prevalent central nervous system nAChR subtype contains α4 and β2 subunits, in two ratios. (α4β2)2β2-nAChR have high agonist sensitivity (HS-isoform), whereas (α4β2)2α4-nAChR agonist responses exhibit a small high-sensitivity, and a predominant low-sensitivity, phase of function (LS-isoform). Multiple non-synonymous mutations in the second and third transmembrane domains of α4 and β2 subunits are associated with SHE. We recently demonstrated that two additional, SHE-associated, missense mutations in the major cytoplasmic loops of these subunits [α4(R336H) and β2(V337G)] cause increased macroscopic function-per receptor. Here, we use single-channel patch-clamp electrophysiology to show that these mutations influence single-channel amplitudes and open- and closed-state kinetics. Pure populations of HS- or LS-isoform α4β2-nAChR were expressed by injecting either 1:10 or 30:1 α4:β2 cRNA ratios, respectively, into Xenopus laevis oocytes. Functional properties of the resulting mutant α4β2-nAChR isoforms were compared to their wildtype counterparts. α4(R336H) subunit incorporation minimally affected single-channel amplitudes, whereas β2(V337G) subunit incorporation reduced them significantly in both isoforms. However, for both mutant subunits, increased function-per-receptor was predominantly caused by altered single channel kinetics. The α4(R336H) mutation primarily destabilizes desensitized states between openings. By contrast, the β2(V337G) mutation principally stabilizes receptor open states. The use of naturally-occurring and physiologically-impactful mutations has allowed us to define valuable new insights regarding the functional roles of nAChR intracellular domains. Further mechanistic context is provided by intracellular-domain structures recently published for other members of the Cys-loop receptor superfamily (α3β4-nAChR and 5-HT3AR).
Collapse
Affiliation(s)
- Maegan M. Weltzin
- Division of Neurobiology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, United States of America
| | - Andrew A. George
- Division of Neurobiology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, United States of America
| | - Ronald J. Lukas
- Division of Neurobiology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, United States of America
| | - Paul Whiteaker
- Division of Neurobiology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, United States of America
| |
Collapse
|
29
|
Al-Omoush TK, Alzoubi KH, Khabour OF, Alsheyab FM, Abu-Siniyeh A, Al-Sawalha NA, Mayyas FA, Cobb CO, Eissenberg T. The CHRNA5 Polymorphism (rs16969968) and its Association with Waterpipe Smoking Addiction among Jordanians. ARAB JOURNAL OF BASIC AND APPLIED SCIENCES 2021; 27:450-455. [PMID: 33511332 DOI: 10.1080/25765299.2020.1849491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Waterpipe smoking is a form of tobacco use that causes nicotine/tobacco dependence and has become a global health problem. In the current study, the association of rs16969968 SNP in the CHRNA5 gene with waterpipe dependence was investigated. A total of 386 men and women who used a waterpipe to smoke tobacco were recruited and divided into less dependent and more dependent smokers based on their score on the Lebanon Waterpipe Dependence Scale (LWDS). Results showed a significant difference in the distribution of GG, GA, and AA genotypes by waterpipe dependence status (P<0.001). The more dependent group showed a higher frequency of the AA genotype than the less dependent smokers' group (38% versus 23% respectively). In addition, the more dependent smokers exhibited more A allele than less dependent smokers (53% versus 37% respectively, P<0.001). In conclusion, there is an association between the rs16969968 SNP and waterpipe dependence as assessed by the LWDS.
Collapse
Affiliation(s)
- Thaka'a K Al-Omoush
- Department of Applied Biology, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H Alzoubi
- Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F Khabour
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Fawzi M Alsheyab
- Department of Applied Biology, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, Jordan
| | - Ahmed Abu-Siniyeh
- Department of Medical Laboratories, Faculty of Health Sciences, American University of Madaba, Madaba, Jordan
| | - Nour A Al-Sawalha
- Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Fadia A Mayyas
- Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Caroline O Cobb
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA.,Center for the Study of Tobacco Products, Virginia Commonwealth University, Richmond, VA, USA
| | - Thomas Eissenberg
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA.,Center for the Study of Tobacco Products, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
30
|
Hopkins RJ, Duan F, Gamble GD, Chiles C, Cavadino A, Billings P, Aberle D, Young RP. Chr15q25 genetic variant (rs16969968) independently confers risk of lung cancer, COPD and smoking intensity in a prospective study of high-risk smokers. Thorax 2021; 76:272-280. [PMID: 33419953 DOI: 10.1136/thoraxjnl-2020-214839] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 10/27/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022]
Abstract
IMPORTANCE While cholinergic receptor nicotinic alpha 5 (CHRNA5) variants have been linked to lung cancer, chronic obstructive pulmonary disease (COPD) and smoking addiction in case-controls studies, their corelationship is not well understood and requires retesting in a cohort study. OBJECTIVE To re-examine the association between the CHRNA5 variant (rs16969968 AA genotype) and the development of lung cancer, relative to its association with COPD and smoking. METHODS In 9270 Non-Hispanic white subjects from the National Lung Screening Trial, a substudy of high-risk smokers were followed for an average of 6.4 years. We compared CHRNA5 genotype according to baseline smoking exposure, lung function and COPD status. We also compared the lung cancer incidence rate, and used multiple logistic regression and mediation analysis to examine the role of the AA genotype of the CHRNA5 variant in smoking exposure, COPD and lung cancer. RESULTS As previously reported, we found the AA high-risk genotype was associated with lower lung function (p=0.005), greater smoking intensity (p<0.001), the presence of COPD (OR 1.28 (95% CI 1.10 to 1.49) p=0.0015) and the development of lung cancer (HR 1.41, (95% CI 1.03 to 1.93) p=0.03). In a mediation analyses, the AA genotype was independently associated with smoking intensity (OR 1.42 (95% CI 1.25 to 1.60, p<0.0001), COPD (OR 1.25, (95% CI 1.66 to 2.53), p=0.0015) and developing lung cancer (OR 1.37, (95% CI 1.03 to 1.82) p=0.03). CONCLUSION In this large-prospective study, we found the CHRNA5 rs 16 969 968 AA genotype to be independently associated with smoking exposure, COPD and lung cancer (triple whammy effect).
Collapse
Affiliation(s)
- Raewyn J Hopkins
- The University of Auckland Faculty of Medical and Health Sciences, Auckland, New Zealand
| | - Fenghai Duan
- Department of Biostatistics and Centre for Statistical Science, Brown University, Providence, Rhode Island, USA
| | - Greg D Gamble
- The University of Auckland Faculty of Medical and Health Sciences, Auckland, New Zealand
| | - Caroline Chiles
- Department of Radiology, Wake Forest Baptist Medical Comprehensive Cancer Center, Winston-Salem, North Carolina, USA
| | - Alana Cavadino
- The University of Auckland Faculty of Medical and Health Sciences, Auckland, New Zealand
| | | | - Denise Aberle
- Department of Radiological Sciences, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California, USA
| | - Robert P Young
- The University of Auckland Faculty of Medical and Health Sciences, Auckland, New Zealand
| |
Collapse
|
31
|
Scholze P, Huck S. The α5 Nicotinic Acetylcholine Receptor Subunit Differentially Modulates α4β2 * and α3β4 * Receptors. Front Synaptic Neurosci 2020; 12:607959. [PMID: 33343327 PMCID: PMC7744819 DOI: 10.3389/fnsyn.2020.607959] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/11/2020] [Indexed: 11/30/2022] Open
Abstract
Nicotine, the principal reinforcing compound in tobacco, acts in the brain by activating neuronal nicotinic acetylcholine receptors (nAChRs). This review summarizes our current knowledge regarding how the α5 accessory nAChR subunit, encoded by the CHRNA5 gene, differentially modulates α4β2* and α3β4* receptors at the cellular level. Genome-wide association studies have linked a gene cluster in chromosomal region 15q25 to increased susceptibility to nicotine addiction, lung cancer, chronic obstructive pulmonary disease, and peripheral arterial disease. Interestingly, this gene cluster contains a non-synonymous single-nucleotide polymorphism (SNP) in the human CHRNA5 gene, causing an aspartic acid (D) to asparagine (N) substitution at amino acid position 398 in the α5 nAChR subunit. Although other SNPs have been associated with tobacco smoking behavior, efforts have focused predominantly on the D398 and N398 variants in the α5 subunit. In recent years, significant progress has been made toward understanding the role that the α5 nAChR subunit—and the role of the D398 and N398 variants—plays on nAChR function at the cellular level. These insights stem primarily from a wide range of experimental models, including receptors expressed heterologously in Xenopus oocytes, various cell lines, and neurons derived from human induced pluripotent stem cells (iPSCs), as well as endogenous receptors in genetically engineered mice and—more recently—rats. Despite providing a wealth of available data, however, these studies have yielded conflicting results, and our understanding of the modulatory role that the α5 subunit plays remains incomplete. Here, we review these reports and the various techniques used for expression and analysis in order to examine how the α5 subunit modulates key functions in α4β2* and α3β4* receptors, including receptor trafficking, sensitivity, efficacy, and desensitization. In addition, we highlight the strikingly different role that the α5 subunit plays in Ca2+ signaling between α4β2* and α3β4* receptors, and we discuss whether the N398 α5 subunit variant can partially replace the D398 variant.
Collapse
Affiliation(s)
- Petra Scholze
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Sigismund Huck
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
32
|
Nadalin S, Flego V, Pavlić SD, Volarić D, Radojčić Badovinac A, Kapović M, Ristić S. Association between the ACE-I/D polymorphism and nicotine dependence amongst patients with lung cancer. Biomed Rep 2020; 13:58. [PMID: 33123372 DOI: 10.3892/br.2020.1365] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/14/2020] [Indexed: 11/06/2022] Open
Abstract
The biologically active peptide angiotensin II is cleaved from angiotensinogen by the renin and the angiotensin-converting enzyme (ACE), an enzymatic cascade known as the renin-angiotensin system (RAS). RAS may be important in the etiology of nicotine dependence by influencing dopaminergic signaling. In the present study, the association between an insertion/deletion (I/D) polymorphism of ACE and nicotine dependence amongst patients with lung cancer was assessed. To date, several studies have shown the relevance of this polymorphic variant in both nicotine dependence and lung cancer. However, the present study is the first to address the potential role of the ACE-I/D polymorphism in nicotine dependence among patients with lung cancer. Genotyping was performed in 305 patients with lung cancer (males/females, 214/91). Significantly more male smokers had the ACE-I allele compared with male non-smokers (44.9 vs. 20.0%; P<0.05). The risk of smoking was ~5-fold higher for males with the ACE-I allele (ACE-II homozygous and ACE-ID heterozygous) vs. ACE-DD homozygous (odds ratio, 5.47; 95% confidence interval, 1.4-21.9; P=0.016). The pack-year smoking history in a subgroup of females with squamous cell carcinoma carrying the ACE-I allele was significantly lower compared with ACE-DD (37.1±14.1 vs. 57.0±29.1; F=4.5; P=0.046). The ACE-I/D polymorphism accounted for 17.6% of the smoking severity in this patient group (β, -0.42; multiple R2 change, 0.176; P=0.046). These results suggest that the ACE-I/D polymorphism contributes to the risk of nicotine dependence and smoking severity in lung cancer patients in a sex-specific manner.
Collapse
Affiliation(s)
- Sergej Nadalin
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Veljko Flego
- Department of Pulmonology, Clinical Hospital Center Rijeka, University of Rijeka, 51000 Rijeka, Croatia
| | - Sanja Dević Pavlić
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Darian Volarić
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Anđelka Radojčić Badovinac
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia.,Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - Miljenko Kapović
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Smiljana Ristić
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
33
|
Abstract
Canalization refers to the evolution of populations such that the number of individuals who deviate from the optimum trait, or experience disease, is minimized. In the presence of rapid cultural, environmental, or genetic change, the reverse process of decanalization may contribute to observed increases in disease prevalence. This review starts by defining relevant concepts, drawing distinctions between the canalization of populations and robustness of individuals. It then considers evidence pertaining to three continuous traits and six domains of disease. In each case, existing genetic evidence for genotype-by-environment interactions is insufficient to support a strong inference of decanalization, but we argue that the advent of genome-wide polygenic risk assessment now makes an empirical evaluation of the role of canalization in preventing disease possible. Finally, the contributions of both rare and common variants to congenital abnormality and adult onset disease are considered in light of a new kerplunk model of genetic effects.
Collapse
Affiliation(s)
- Greg Gibson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA;
| | - Kristine A Lacek
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA;
| |
Collapse
|
34
|
Icick R, Forget B, Cloëz-Tayarani I, Pons S, Maskos U, Besson M. Genetic susceptibility to nicotine addiction: Advances and shortcomings in our understanding of the CHRNA5/A3/B4 gene cluster contribution. Neuropharmacology 2020; 177:108234. [PMID: 32738310 DOI: 10.1016/j.neuropharm.2020.108234] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/28/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
Over the last decade, robust human genetic findings have been instrumental in elucidating the heritable basis of nicotine addiction (NA). They highlight coding and synonymous polymorphisms in a cluster on chromosome 15, encompassing the CHRNA5, CHRNA3 and CHRNB4 genes, coding for three subunits of the nicotinic acetylcholine receptor (nAChR). They have inspired an important number of preclinical studies, and will hopefully lead to the definition of novel drug targets for treating NA. Here, we review these candidate gene and genome-wide association studies (GWAS) and their direct implication in human brain function and NA-related phenotypes. We continue with a description of preclinical work in transgenic rodents that has led to a mechanistic understanding of several of the genetic hits. We also highlight important issues with regards to CHRNA3 and CHRNB4 where we are still lacking a dissection of their role in NA, including even in preclinical models. We further emphasize the use of human induced pluripotent stem cell-derived models for the analysis of synonymous and intronic variants on a human genomic background. Finally, we indicate potential avenues to further our understanding of the role of this human genetic variation. This article is part of the special issue on 'Contemporary Advances in Nicotine Neuropharmacology'.
Collapse
Affiliation(s)
- Romain Icick
- Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR3571, Institut Pasteur, 25 Rue du Dr Roux, 75724, Paris Cedex 15, France; Département de Psychiatrie et de Médecine Addictologique, Groupe Hospitalier Saint-Louis, Lariboisière, Fernand Widal, Assistance-Publique Hôpitaux de Paris, Paris, F-75010, France; INSERM UMR-S1144, Paris, F-75006, France; FHU "NOR-SUD", Assistance-Publique Hôpitaux de Paris, Paris, F-75001, France
| | - Benoît Forget
- Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR3571, Institut Pasteur, 25 Rue du Dr Roux, 75724, Paris Cedex 15, France; Génétique Humaine et Fonctions Cognitives, CNRS UMR3571, Institut Pasteur, 25 Rue du Dr Roux, 75724, Paris Cedex 15, France
| | - Isabelle Cloëz-Tayarani
- Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR3571, Institut Pasteur, 25 Rue du Dr Roux, 75724, Paris Cedex 15, France; FHU "NOR-SUD", Assistance-Publique Hôpitaux de Paris, Paris, F-75001, France
| | - Stéphanie Pons
- Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR3571, Institut Pasteur, 25 Rue du Dr Roux, 75724, Paris Cedex 15, France; FHU "NOR-SUD", Assistance-Publique Hôpitaux de Paris, Paris, F-75001, France
| | - Uwe Maskos
- Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR3571, Institut Pasteur, 25 Rue du Dr Roux, 75724, Paris Cedex 15, France; FHU "NOR-SUD", Assistance-Publique Hôpitaux de Paris, Paris, F-75001, France
| | - Morgane Besson
- Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR3571, Institut Pasteur, 25 Rue du Dr Roux, 75724, Paris Cedex 15, France; FHU "NOR-SUD", Assistance-Publique Hôpitaux de Paris, Paris, F-75001, France.
| |
Collapse
|
35
|
Panagiotou OA, Schuit E, Munafò MR, Bennett DA, Bergen AW, David SP. Smoking Cessation Pharmacotherapy Based on Genetically-Informed Biomarkers: What is the Evidence? Nicotine Tob Res 2020; 21:1289-1293. [PMID: 30690475 DOI: 10.1093/ntr/ntz009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 01/17/2019] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Pharmacogenomic studies have used genetic variants to identify smokers likely to respond to pharmacological treatments for smoking cessation. METHODS We performed a systematic review and meta-analysis of primary and secondary analyses of trials of smoking cessation pharmacotherapies. Eligible were trials with data on a priori selected single nucleotide polymorphisms, replicated non-single nucleotide polymorphisms, and/or the nicotine metabolite ratio. We estimated the genotype × treatment interaction as the ratio of risk ratios (RRR) for treatment effects across genotype groups. RESULTS We identified 18 trials (N = 9017 participants), including 40 active (bupropion, nicotine replacement therapy [NRT], varenicline, or combination therapies) versus placebo comparisons and 16 active versus active comparisons. There was statistical evidence of heterogeneity across rs16969968 genotypes in CHRNA5 with regard to both 6-month abstinence and end-of-treatment abstinence in non-Hispanic black smokers and end-of-treatment abstinence in non-Hispanic white smokers. There was also heterogeneity across rs1051730 genotypes in CHRNA3 with regard to end-of-treatment abstinence in non-Hispanic white smokers. There was no clear statistical evidence for other genotype-by-treatment combinations. Compared with placebo, NRT was more effective among non-Hispanic black smokers with rs16969968-GG with regard to both 6-month abstinence (RRR for GG vs. GA or AA, 3.51; 95% confidence interval [CI] = 1.19 to 10.30) and end-of-treatment abstinence (RRR for GG vs. GA or AA, 5.84; 95% CI = 1.89 to 18.10). Among non-Hispanic white smokers, NRT effectiveness relative to placebo was comparable across rs1051730 and rs169969960 genotypes. CONCLUSIONS We did not identify widespread differential effects of smoking cessation pharmacotherapies based on genotype. The quality of the evidence is generally moderate. IMPLICATIONS Although we identified some evidence of genotype × treatment interactions, the vast majority of analyses did not provide evidence of differential treatment response by genotype. Where we find some evidence, these results should be considered preliminary and interpreted with caution because of the small number of contributing trials per genotype comparison, the wide confidence intervals, and the moderate quality of evidence. Prospective trials and individual-patient data meta-analyses accounting for heterogeneity of treatment effects through modeling are needed to assess the clinical utility of genetically informed biomarkers to guide pharmacotherapy choice for smoking cessation.
Collapse
Affiliation(s)
- Orestis A Panagiotou
- Department of Health Services, Policy and Practice, Brown University School of Public Health, Providence, RI.,Center for Gerontology and Healthcare Research, Brown University School of Public Health, Providence, RI
| | - Ewoud Schuit
- Cochrane Netherlands, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA
| | - Marcus R Munafò
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,School of Psychological Science, University of Bristol, Bristol, UK
| | - Derrick A Bennett
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Andrew W Bergen
- Biorealm, LLC, Walnut, CA.,Oregon Research Institute, Eugene, OR
| | - Sean P David
- Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA.,Division of Primary Care and Population Health, Department of Medicine, Stanford University, Stanford, CA
| |
Collapse
|
36
|
Cross SJ, Reynaga DD, Cano M, Belluzzi JD, Zaveri NT, Leslie FM. Differences in mechanisms underlying reinstatement of cigarette smoke extract- and nicotine-seeking behavior in rats. Neuropharmacology 2020; 162:107846. [PMID: 31704271 PMCID: PMC7034132 DOI: 10.1016/j.neuropharm.2019.107846] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 10/18/2019] [Accepted: 11/05/2019] [Indexed: 12/28/2022]
Abstract
Despite extensive research, current therapies for smoking cessation are largely ineffective at maintaining abstinence for more than a year. Whereas most preclinical studies use nicotine alone, the goal of the present study was to evaluate whether inclusion of non-nicotine tobacco constituents provides better face validity for the development of new pharmacological therapies for smoking cessation. Here, we trained adult male rats to self-administer nicotine alone or cigarette smoke extract (CSE), which contains nicotine and other aqueous constituents of cigarette smoke. After stable self-administration behavior was established, animals underwent extinction training followed by drug and cue primed reinstatement testing. We show that animals that self-administered CSE had significant reinstatement in all drug and drug + cue stimulus conditions whereas animals that self-administered nicotine only showed significant reinstatement in the drug + cue conditions. AT-1001, an α3β4 nicotinic acetylcholine receptor (nAChR) functional antagonist, attenuated drug + cue-primed reinstatement of both CSE- and nicotine-seeking behavior. However, AT-1001 was less potent in blocking drug-primed reinstatement in animals that had self-administered CSE than in those that had self-administered nicotine alone. This was the case even when nicotine was used to prime reinstatement in animals that had self-administered CSE, suggesting that prior CSE exposure had altered the functional role of α3β4-containing nAChRs in drug-seeking behavior. These findings confirm the importance of non-nicotine tobacco constituents and α3β4* nAChRs in cue- and nicotine-primed craving. They also suggest that tests using CSE may be more valid models to study tobacco dependence than use of nicotine alone.
Collapse
Affiliation(s)
- Sarah J Cross
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA.
| | - Daisy D Reynaga
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Michelle Cano
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - James D Belluzzi
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | | | - Frances M Leslie
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA; Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| |
Collapse
|
37
|
Santoro A, Tomino C, Prinzi G, Lamonaca P, Cardaci V, Fini M, Russo P. Tobacco Smoking: Risk to Develop Addiction, Chronic Obstructive Pulmonary Disease, and Lung Cancer. Recent Pat Anticancer Drug Discov 2019; 14:39-52. [PMID: 30605063 DOI: 10.2174/1574892814666190102122848] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/11/2018] [Accepted: 12/27/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND The morbidity and mortality associated with tobacco smoking is well established. Nicotine is the addictive component of tobacco. Nicotine, through the non-neuronal α7nicotinic receptor, induces cell proliferation, neo-angiogenesis, epithelial to mesenchymal transition, and inhibits drug-induced apoptosis. OBJECTIVE To understand the genetic, molecular and cellular biology of addiction, chronic obstructive pulmonary disease and lung cancer. METHODS The search for papers to be included in the review was performed during the months of July- September 2018 in the following databases: PubMed (http://www.ncbi.nlm.nih.gov), Scopus (http://www.scopus.com), EMBASE (http://www.elsevier.com/online-tools/embase), and ISI Web of Knowledge (http://apps.webofknowledge.com/). The following searching terms: "nicotine", "nicotinic receptor", and "addiction" or "COPD" or "lung cancer" were used. Patents were retrieved in clinicaltrials.gov (https://clinicaltrials.gov/). All papers written in English were evaluated. The reference list of retrieved articles was also reviewed to identify other eligible studies that were not indexed by the above-mentioned databases. New experimental data on the ability of nicotine to promote transformation of human bronchial epithelial cells, exposed for one hour to Benzo[a]pyrene-7,8-diol-9-10-epoxide, are reported. RESULTS Nicotinic receptors variants and nicotinic receptors upregulation are involved in addiction, chronic obstructive pulmonary disease and/or lung cancer. Nicotine through α7nicotinic receptor upregulation induces complete bronchial epithelial cells transformation. CONCLUSION Genetic studies highlight the involvement of nicotinic receptors variants in addiction, chronic obstructive pulmonary disease and/or lung cancer. A future important step will be to translate these genetic findings to clinical practice. Interventions able to help smoking cessation in nicotine dependence subjects, under patent, are reported.
Collapse
Affiliation(s)
- Alessia Santoro
- Clinical and Molecular Epidemiology, IRCSS San Raffaele Pisana, Via di Valcannuta 247, I-00166 Rome, Italy
| | - Carlo Tomino
- Scientific Direction, IRCSS San Raffaele Pisana, Via di Valcannuta 247, I-00166 Rome, Italy
| | - Giulia Prinzi
- Clinical and Molecular Epidemiology, IRCSS San Raffaele Pisana, Via di Valcannuta 247, I-00166 Rome, Italy
| | - Palma Lamonaca
- Clinical and Molecular Epidemiology, IRCSS San Raffaele Pisana, Via di Valcannuta 247, I-00166 Rome, Italy
| | - Vittorio Cardaci
- Pulmonary Rehabilitation, IRCCS San Raffaele Pisana, Via della Pisana, 235, I-00163 Rome, Italy
| | - Massimo Fini
- Scientific Direction, IRCSS San Raffaele Pisana, Via di Valcannuta 247, I-00166 Rome, Italy
| | - Patrizia Russo
- Clinical and Molecular Epidemiology, IRCSS San Raffaele Pisana, Via di Valcannuta 247, I-00166 Rome, Italy
| |
Collapse
|
38
|
Bi W, Zhao Z, Dey R, Fritsche LG, Mukherjee B, Lee S. A Fast and Accurate Method for Genome-wide Scale Phenome-wide G × E Analysis and Its Application to UK Biobank. Am J Hum Genet 2019; 105:1182-1192. [PMID: 31735295 PMCID: PMC6904814 DOI: 10.1016/j.ajhg.2019.10.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023] Open
Abstract
The etiology of most complex diseases involves genetic variants, environmental factors, and gene-environment interaction (G × E) effects. Compared with marginal genetic association studies, G × E analysis requires more samples and detailed measure of environmental exposures, and this limits the possible discoveries. Large-scale population-based biobanks with detailed phenotypic and environmental information, such as UK-Biobank, can be ideal resources for identifying G × E effects. However, due to the large computation cost and the presence of case-control imbalance, existing methods often fail. Here we propose a scalable and accurate method, SPAGE (SaddlePoint Approximation implementation of G × E analysis), that is applicable for genome-wide scale phenome-wide G × E studies. SPAGE fits a genotype-independent logistic model only once across the genome-wide analysis in order to reduce computation cost, and SPAGE uses a saddlepoint approximation (SPA) to calibrate the test statistics for analysis of phenotypes with unbalanced case-control ratios. Simulation studies show that SPAGE is 33-79 times faster than the Wald test and 72-439 times faster than the Firth's test, and SPAGE can control type I error rates at the genome-wide significance level even when case-control ratios are extremely unbalanced. Through the analysis of UK-Biobank data of 344,341 white British European-ancestry samples, we show that SPAGE can efficiently analyze large samples while controlling for unbalanced case-control ratios.
Collapse
Affiliation(s)
- Wenjian Bi
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhangchen Zhao
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rounak Dey
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Lars G Fritsche
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Seunggeun Lee
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
39
|
Langdon RJ, Richmond RC, Hemani G, Zheng J, Wade KH, Carreras-Torres R, Johansson M, Brennan P, Wootton RE, Munafo MR, Smith GD, Relton CL, Vincent EE, Martin RM, Haycock P. A Phenome-Wide Mendelian Randomization Study of Pancreatic Cancer Using Summary Genetic Data. Cancer Epidemiol Biomarkers Prev 2019; 28:2070-2078. [PMID: 31315910 DOI: 10.1158/1055-9965.epi-19-0036] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/29/2019] [Accepted: 07/10/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The 5-year mortality rate for pancreatic cancer is among the highest of all cancers. Greater understanding of underlying causes could inform population-wide intervention strategies for prevention. Summary genetic data from genome-wide association studies (GWAS) have become available for thousands of phenotypes. These data can be exploited in Mendelian randomization (MR) phenome-wide association studies (PheWAS) to efficiently screen the phenome for potential determinants of disease risk. METHODS We conducted an MR-PheWAS of pancreatic cancer using 486 phenotypes, proxied by 9,124 genetic variants, and summary genetic data from a GWAS of pancreatic cancer (7,110 cancer cases, 7,264 controls). ORs and 95% confidence intervals per 1 SD increase in each phenotype were generated. RESULTS We found evidence that previously reported risk factors of body mass index (BMI; 1.46; 1.20-1.78) and hip circumference (1.42; 1.21-1.67) were associated with pancreatic cancer. We also found evidence of novel associations with metabolites that have not previously been implicated in pancreatic cancer: ADpSGEGDFXAEGGGVR*, a fibrinogen-cleavage peptide (1.60; 1.31-1.95), and O-sulfo-l-tyrosine (0.58; 0.46-0.74). An inverse association was also observed with lung adenocarcinoma (0.63; 0.54-0.74). CONCLUSIONS Markers of adiposity (BMI and hip circumference) are potential intervention targets for pancreatic cancer prevention. Further clarification of the causal relevance of the fibrinogen-cleavage peptides and O-sulfo-l-tyrosine in pancreatic cancer etiology is required, as is the basis of our observed association with lung adenocarcinoma. IMPACT For pancreatic cancer, MR-PheWAS can augment existing risk factor knowledge and generate novel hypotheses to investigate.
Collapse
Affiliation(s)
- Ryan J Langdon
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Rebecca C Richmond
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Gibran Hemani
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Jie Zheng
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Kaitlin H Wade
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Robert Carreras-Torres
- Biomarkers and Susceptibility Unit, IDIBELL-Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Mattias Johansson
- Section of Genetics, International Agency for Research on Cancer (IARC), Lyon, France
| | - Paul Brennan
- Section of Genetics, International Agency for Research on Cancer (IARC), Lyon, France
| | - Robyn E Wootton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, United Kingdom
- UK Centre for Tobacco and Alcohol Studies, School of Experimental Psychology, University of Bristol, Bristol, United Kingdom
| | - Marcus R Munafo
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, United Kingdom
- UK Centre for Tobacco and Alcohol Studies, School of Experimental Psychology, University of Bristol, Bristol, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Emma E Vincent
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Richard M Martin
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Philip Haycock
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom.
- Bristol Medical School, Department of Population Health Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
40
|
Yuan X, Biswas S. Bivariate logistic Bayesian LASSO for detecting rare haplotype association with two correlated phenotypes. Genet Epidemiol 2019; 43:996-1017. [PMID: 31544985 DOI: 10.1002/gepi.22258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/31/2019] [Accepted: 08/09/2019] [Indexed: 11/08/2022]
Abstract
In genetic association studies, joint modeling of related traits/phenotypes can utilize the correlation between them and thereby provide more power and uncover additional information about genetic etiology. Moreover, detecting rare genetic variants are of current scientific interest as a key to missing heritability. Logistic Bayesian LASSO (LBL) has been proposed recently to detect rare haplotype variants using case-control data, that is, a single binary phenotype. As there is currently no haplotype association method that can handle multiple binary phenotypes, we extend LBL to fill this gap. We develop a bivariate model by using a latent variable to induce correlation between the two outcomes. We carry out extensive simulations to investigate the bivariate LBL and compare with the univariate LBL. The bivariate LBL performs better or similar to the univariate LBL in most settings. It has the highest gain in power when a haplotype is associated with both traits and it affects at least one trait in a direction opposite to the direction of the correlation between the traits. We analyze two data sets-Genetic Analysis Workshop 19 sequence data on systolic and diastolic blood pressures and a genome-wide association data set on lung cancer and smoking and detect several associated rare haplotypes.
Collapse
Affiliation(s)
- Xiaochen Yuan
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, Texas
| | - Swati Biswas
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, Texas
| |
Collapse
|
41
|
Gu F, Zhao C, Jiang T, Li X, Mao Y, Zhou C. Association Between Nicotine-dependent Gene Polymorphism and Smoking Cessation in Patients With Lung Cancer. Clin Lung Cancer 2019; 21:171-176. [PMID: 31402126 DOI: 10.1016/j.cllc.2019.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/31/2019] [Accepted: 07/09/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Patients with lung cancer continue to smoke owing to complex factors. Failure to quit smoking (defined as nicotine dependence) is significantly associated with genetic status. This study aimed to investigate the relationship between polymorphisms in nicotine dependence genes and smoking status after the diagnosis of lung cancer. PATIENTS AND METHODS A total of 240 patients with lung cancer were included from July 2017 to March 2018. According to the actual smoking condition after lung cancer diagnosis, eligible patients were divided into 3 groups: the never-smoking group, the failure to quit smoking group, and the successful smoking cessation group. Fagerstrom Test for Nicotine Dependence scores were used to evaluate the smoking status of each group. Three nicotine-dependent genes with 6 loci were detected. RESULTS Among the 240 patients, 86 were never-smokers, 51 failed to quit smoking, and 104 successfully quit smoking. The initial age of smoking in the failure to quit smoking group was significantly younger than those in the successful smoking cessation group (P = .001). There was a significant difference in the GG and AG and AA genotype distributions of CHRNA3 (rs578776) among the 3 groups (P = .003). There was also a significant difference in the distribution of CHRNA4 (rs2229959) genotypes among the 3 groups (P = .003). However, there was no significant difference in the genotype distribution of CHRNA5 (rs588765) among the 3 groups (P = .277). CONCLUSIONS Gene polymorphisms of CHRNA3 (rs578776) and CHRNA4 (rs1044396 and rs2229959) were associated with the success of smoking cessation after the diagnosis of lung cancer, which should be considered in the management of smoking cessation after patients are diagnosed with lung cancer.
Collapse
Affiliation(s)
- Fen Gu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chao Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuefei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanjun Mao
- Nursing Department, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
42
|
Brazel DM, Jiang Y, Hughey JM, Turcot V, Zhan X, Gong J, Batini C, Weissenkampen JD, Liu M, Barnes DR, Bertelsen S, Chou YL, Erzurumluoglu AM, Faul JD, Haessler J, Hammerschlag AR, Hsu C, Kapoor M, Lai D, Le N, de Leeuw CA, Loukola A, Mangino M, Melbourne CA, Pistis G, Qaiser B, Rohde R, Shao Y, Stringham H, Wetherill L, Zhao W, Agrawal A, Bierut L, Chen C, Eaton CB, Goate A, Haiman C, Heath A, Iacono WG, Martin NG, Polderman TJ, Reiner A, Rice J, Schlessinger D, Scholte HS, Smith JA, Tardif JC, Tindle HA, van der Leij AR, Boehnke M, Chang-Claude J, Cucca F, David SP, Foroud T, Howson JMM, Kardia SLR, Kooperberg C, Laakso M, Lettre G, Madden P, McGue M, North K, Posthuma D, Spector T, Stram D, Tobin MD, Weir DR, Kaprio J, Abecasis GR, Liu DJ, Vrieze S. Exome Chip Meta-analysis Fine Maps Causal Variants and Elucidates the Genetic Architecture of Rare Coding Variants in Smoking and Alcohol Use. Biol Psychiatry 2019; 85:946-955. [PMID: 30679032 PMCID: PMC6534468 DOI: 10.1016/j.biopsych.2018.11.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/05/2018] [Accepted: 11/29/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Smoking and alcohol use have been associated with common genetic variants in multiple loci. Rare variants within these loci hold promise in the identification of biological mechanisms in substance use. Exome arrays and genotype imputation can now efficiently genotype rare nonsynonymous and loss of function variants. Such variants are expected to have deleterious functional consequences and to contribute to disease risk. METHODS We analyzed ∼250,000 rare variants from 16 independent studies genotyped with exome arrays and augmented this dataset with imputed data from the UK Biobank. Associations were tested for five phenotypes: cigarettes per day, pack-years, smoking initiation, age of smoking initiation, and alcoholic drinks per week. We conducted stratified heritability analyses, single-variant tests, and gene-based burden tests of nonsynonymous/loss-of-function coding variants. We performed a novel fine-mapping analysis to winnow the number of putative causal variants within associated loci. RESULTS Meta-analytic sample sizes ranged from 152,348 to 433,216, depending on the phenotype. Rare coding variation explained 1.1% to 2.2% of phenotypic variance, reflecting 11% to 18% of the total single nucleotide polymorphism heritability of these phenotypes. We identified 171 genome-wide associated loci across all phenotypes. Fine mapping identified putative causal variants with double base-pair resolution at 24 of these loci, and between three and 10 variants for 65 loci. Twenty loci contained rare coding variants in the 95% credible intervals. CONCLUSIONS Rare coding variation significantly contributes to the heritability of smoking and alcohol use. Fine-mapping genome-wide association study loci identifies specific variants contributing to the biological etiology of substance use behavior.
Collapse
Affiliation(s)
- David M Brazel
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado; Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado
| | - Yu Jiang
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Jordan M Hughey
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Valérie Turcot
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Montreal, Quebec, Canada
| | - Xiaowei Zhan
- Department of Clinical Science, Center for Genetics of Host Defense, University of Texas Southwestern, Dallas, Texas
| | - Jian Gong
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Chiara Batini
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - J Dylan Weissenkampen
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - MengZhen Liu
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota
| | - Daniel R Barnes
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Sarah Bertelsen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yi-Ling Chou
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | | | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan
| | - Jeff Haessler
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Anke R Hammerschlag
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Chris Hsu
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Manav Kapoor
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Dongbing Lai
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Nhung Le
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois
| | - Christiaan A de Leeuw
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Anu Loukola
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland; Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom; National Institute for Health Research Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, United Kingdom
| | - Carl A Melbourne
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Giorgio Pistis
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | - Beenish Qaiser
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland; Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Rebecca Rohde
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Yaming Shao
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Heather Stringham
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Leah Wetherill
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Laura Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Chu Chen
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Epidemiology, Head and Neck Surgery Center, University of Washington, Seattle, Washington; Department of Otolaryngology, Head and Neck Surgery Center, University of Washington, Seattle, Washington
| | - Charles B Eaton
- Department of Family Medicine, Brown University, Providence, Rhode Island
| | - Alison Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Christopher Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Andrew Heath
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - William G Iacono
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota
| | | | - Tinca J Polderman
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Alex Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Epidemiology, Head and Neck Surgery Center, University of Washington, Seattle, Washington
| | - John Rice
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri; Department of Mathematics, Washington University in St. Louis, St. Louis, Missouri
| | - David Schlessinger
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - H Steven Scholte
- Department of Psychology, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, the Netherlands
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Jean-Claude Tardif
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Montreal, Quebec, Canada
| | - Hilary A Tindle
- Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Andries R van der Leij
- Department of Psychology, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, the Netherlands
| | - Michael Boehnke
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | - Sean P David
- Department of Medicine, Stanford University, Stanford, California
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Joanna M M Howson
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| | - Charles Kooperberg
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Markku Laakso
- Department of Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Guillaume Lettre
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Montreal Heart Institute, Montreal, Quebec, Canada
| | - Pamela Madden
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Matt McGue
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota
| | - Kari North
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, University of Amsterdam, Amsterdam, the Netherlands; Department of Clinical Genetics, VU University Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Timothy Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Daniel Stram
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Martin D Tobin
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland; Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Gonçalo R Abecasis
- Regeneron Pharmaceuticals, Tarrytown, New York; Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Dajiang J Liu
- Institute of Personalized Medicine, Penn State College of Medicine, Hershey, Pennsylvania.
| | - Scott Vrieze
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
43
|
Gibson M, Munafò MR, Taylor AE, Treur JL. Evidence for Genetic Correlations and Bidirectional, Causal Effects Between Smoking and Sleep Behaviors. Nicotine Tob Res 2019; 21:731-738. [PMID: 30365022 PMCID: PMC6528151 DOI: 10.1093/ntr/nty230] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Cigarette smokers are at increased risk of poor sleep behaviors. However, it is largely unknown whether these associations are due to shared (genetic) risk factors and/or causal effects (which may be bidirectional). METHODS We obtained summary-level data of genome-wide association studies of smoking (smoking initiation [n = 74 035], cigarettes per day [n = 38 181], and smoking cessation [n = 41 278]) and sleep behaviors (sleep duration and chronotype, or "morningness" [n = 128 266] and insomnia [n = 113 006]). Using linkage disequilibrium (LD) score regression, we calculated genetic correlations between smoking and sleep behaviors. To investigate causal effects, we employed Mendelian randomization (MR), both with summary-level data and individual-level data (n = 333 581 UK Biobank participants). For MR with summary-level data, individual genetic variants were combined with inverse variance-weighted meta-analysis, weighted median regression, MR-Robust Adjusted Profile Score, and MR Egger methods. RESULTS We found negative genetic correlations between smoking initiation and sleep duration (rg = -.14, 95% CI = -0.26 to -0.01) and smoking cessation and chronotype (rg = -.18, 95% CI = -0.31 to -0.06), and positive genetic correlations between smoking initiation and insomnia (rg = .27, 95% CI = 0.06 to 0.49) and cigarettes per day and insomnia (rg = .15, 95% CI = 0.01 to 0.28). MR provided strong evidence that smoking more cigarettes causally decreases the odds of being a morning person, (RAPS) and weak evidence that insomnia causally increases smoking heaviness and decreases smoking cessation odds. CONCLUSIONS Smoking and sleep behaviors show moderate genetic correlation. Heavier smoking seems to causally affect circadian rhythm and there is some indication that insomnia increases smoking heaviness and hampers cessation. Our findings point to sleep as a potentially interesting smoking treatment target. IMPLICATIONS Using LD score regression, we found evidence that smoking and different sleep behaviors (sleep duration, chronotype (morningness), and insomnia) are moderately genetically correlated-genetic variants associated with less or poorer sleep also increased the odds of smoking (more heavily). MR analyses suggested that heavier smoking causally affects circadian rhythm (decreasing the odds of being a morning person) and there was some indication that insomnia increases smoking heaviness and hampers smoking cessation. Our findings indicate a complex, bidirectional relationship between smoking and sleep behaviors and point to sleep as a potentially interesting smoking treatment target.
Collapse
Affiliation(s)
- Mark Gibson
- School of Experimental Psychology, University of Bristol, Bristol, UK
| | - Marcus R Munafò
- School of Experimental Psychology, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- UK Centre for Tobacco and Alcohol Studies, Bristol, UK
| | - Amy E Taylor
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Biomedical Research Centre, University Hospitals Bristol NHS Foundation Trust and University of Bristol, UK
| | - Jorien L Treur
- School of Experimental Psychology, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| |
Collapse
|
44
|
Hällfors J, Palviainen T, Surakka I, Gupta R, Buchwald J, Raevuori A, Ripatti S, Korhonen T, Jousilahti P, Madden PA, Kaprio J, Loukola A. Genome-wide association study in Finnish twins highlights the connection between nicotine addiction and neurotrophin signaling pathway. Addict Biol 2019. [PMID: 29532581 PMCID: PMC6519128 DOI: 10.1111/adb.12618] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The heritability of nicotine dependence based on family studies is substantial. Nevertheless, knowledge of the underlying genetic architecture remains meager. Our aim was to identify novel genetic variants responsible for interindividual differences in smoking behavior. We performed a genome-wide association study on 1715 ever smokers ascertained from the population-based Finnish Twin Cohort enriched for heavy smoking. Data imputation used the 1000 Genomes Phase I reference panel together with a whole genome sequence-based Finnish reference panel. We analyzed three measures of nicotine addiction-smoking quantity, nicotine dependence and nicotine withdrawal. We annotated all genome-wide significant SNPs for their functional potential. First, we detected genome-wide significant association on 16p12 with smoking quantity (P = 8.5 × 10-9 ), near CLEC19A. The lead-SNP stands 22 kb from a binding site for NF-κB transcription factors, which play a role in the neurotrophin signaling pathway. However, the signal was not replicated in an independent Finnish population-based sample, FINRISK (n = 6763). Second, nicotine withdrawal showed association on 2q21 in an intron of TMEM163 (P = 2.1 × 10-9 ), and on 11p15 (P = 6.6 × 10-8 ) in an intron of AP2A2, and P = 4.2 × 10-7 for a missense variant in MUC6, both involved in the neurotrophin signaling pathway). Third, association was detected on 3p22.3 for maximum number of cigarettes smoked per day (P = 3.1 × 10-8 ) near STAC. Associating CLEC19A and TMEM163 SNPs were annotated to influence gene expression or methylation. The neurotrophin signaling pathway has previously been associated with smoking behavior. Our findings further support the role in nicotine addiction.
Collapse
Affiliation(s)
- Jenni Hällfors
- Institute for Molecular Medicine Finland (FIMM)University of Helsinki Finland
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland (FIMM)University of Helsinki Finland
| | - Ida Surakka
- Institute for Molecular Medicine Finland (FIMM)University of Helsinki Finland
| | - Richa Gupta
- Institute for Molecular Medicine Finland (FIMM)University of Helsinki Finland
| | - Jadwiga Buchwald
- Institute for Molecular Medicine Finland (FIMM)University of Helsinki Finland
| | - Anu Raevuori
- Department of Public HealthUniversity of Helsinki Finland
- Department of Adolescent PsychiatryHelsinki University Central Hospital Finland
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM)University of Helsinki Finland
- Department of Public HealthUniversity of Helsinki Finland
- Wellcome Trust Sanger Institute UK
| | - Tellervo Korhonen
- Institute for Molecular Medicine Finland (FIMM)University of Helsinki Finland
- Institute of Public Health and Clinical NutritionUniversity of Eastern Finland Finland
| | | | - Pamela A.F. Madden
- Department of PsychiatryWashington University School of Medicine Saint Louis MO USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM)University of Helsinki Finland
- Department of Public HealthUniversity of Helsinki Finland
| | - Anu Loukola
- Institute for Molecular Medicine Finland (FIMM)University of Helsinki Finland
| |
Collapse
|
45
|
Friedman JR, Richbart SD, Merritt JC, Brown KC, Nolan NA, Akers AT, Lau JK, Robateau ZR, Miles SL, Dasgupta P. Acetylcholine signaling system in progression of lung cancers. Pharmacol Ther 2019; 194:222-254. [PMID: 30291908 PMCID: PMC6348061 DOI: 10.1016/j.pharmthera.2018.10.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The neurotransmitter acetylcholine (ACh) acts as an autocrine growth factor for human lung cancer. Several lines of evidence show that lung cancer cells express all of the proteins required for the uptake of choline (choline transporter 1, choline transporter-like proteins) synthesis of ACh (choline acetyltransferase, carnitine acetyltransferase), transport of ACh (vesicular acetylcholine transport, OCTs, OCTNs) and degradation of ACh (acetylcholinesterase, butyrylcholinesterase). The released ACh binds back to nicotinic (nAChRs) and muscarinic receptors on lung cancer cells to accelerate their proliferation, migration and invasion. Out of all components of the cholinergic pathway, the nAChR-signaling has been studied the most intensely. The reason for this trend is due to genome-wide data studies showing that nicotinic receptor subtypes are involved in lung cancer risk, the relationship between cigarette smoke and lung cancer risk as well as the rising popularity of electronic cigarettes considered by many as a "safe" alternative to smoking. There are a small number of articles which review the contribution of the other cholinergic proteins in the pathophysiology of lung cancer. The primary objective of this review article is to discuss the function of the acetylcholine-signaling proteins in the progression of lung cancer. The investigation of the role of cholinergic network in lung cancer will pave the way to novel molecular targets and drugs in this lethal malignancy.
Collapse
Affiliation(s)
- Jamie R Friedman
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Stephen D Richbart
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Justin C Merritt
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Kathleen C Brown
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Nicholas A Nolan
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Austin T Akers
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Jamie K Lau
- Biology Department, Center for the Sciences, Box 6931, Radford University, Radford, Virginia 24142
| | - Zachary R Robateau
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Sarah L Miles
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755
| | - Piyali Dasgupta
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, 1700 Third Avenue, Huntington, WV 25755.
| |
Collapse
|
46
|
Liu M, Jiang Y, Wedow R, Li Y, Brazel DM, Chen F, Datta G, Davila-Velderrain J, McGuire D, Tian C, Zhan X, Choquet H, Docherty AR, Faul JD, Foerster JR, Fritsche LG, Gabrielsen ME, Gordon SD, Haessler J, Hottenga JJ, Huang H, Jang SK, Jansen PR, Ling Y, Mägi R, Matoba N, McMahon G, Mulas A, Orrù V, Palviainen T, Pandit A, Reginsson GW, Skogholt AH, Smith JA, Taylor AE, Turman C, Willemsen G, Young H, Young KA, Zajac GJM, Zhao W, Zhou W, Bjornsdottir G, Boardman JD, Boehnke M, Boomsma DI, Chen C, Cucca F, Davies GE, Eaton CB, Ehringer MA, Esko T, Fiorillo E, Gillespie NA, Gudbjartsson DF, Haller T, Harris KM, Heath AC, Hewitt JK, Hickie IB, Hokanson JE, Hopfer CJ, Hunter DJ, Iacono WG, Johnson EO, Kamatani Y, Kardia SLR, Keller MC, Kellis M, Kooperberg C, Kraft P, Krauter KS, Laakso M, Lind PA, Loukola A, Lutz SM, Madden PAF, Martin NG, McGue M, McQueen MB, Medland SE, Metspalu A, Mohlke KL, Nielsen JB, Okada Y, Peters U, Polderman TJC, Posthuma D, Reiner AP, Rice JP, Rimm E, Rose RJ, Runarsdottir V, Stallings MC, Stančáková A, Stefansson H, Thai KK, Tindle HA, Tyrfingsson T, Wall TL, et alLiu M, Jiang Y, Wedow R, Li Y, Brazel DM, Chen F, Datta G, Davila-Velderrain J, McGuire D, Tian C, Zhan X, Choquet H, Docherty AR, Faul JD, Foerster JR, Fritsche LG, Gabrielsen ME, Gordon SD, Haessler J, Hottenga JJ, Huang H, Jang SK, Jansen PR, Ling Y, Mägi R, Matoba N, McMahon G, Mulas A, Orrù V, Palviainen T, Pandit A, Reginsson GW, Skogholt AH, Smith JA, Taylor AE, Turman C, Willemsen G, Young H, Young KA, Zajac GJM, Zhao W, Zhou W, Bjornsdottir G, Boardman JD, Boehnke M, Boomsma DI, Chen C, Cucca F, Davies GE, Eaton CB, Ehringer MA, Esko T, Fiorillo E, Gillespie NA, Gudbjartsson DF, Haller T, Harris KM, Heath AC, Hewitt JK, Hickie IB, Hokanson JE, Hopfer CJ, Hunter DJ, Iacono WG, Johnson EO, Kamatani Y, Kardia SLR, Keller MC, Kellis M, Kooperberg C, Kraft P, Krauter KS, Laakso M, Lind PA, Loukola A, Lutz SM, Madden PAF, Martin NG, McGue M, McQueen MB, Medland SE, Metspalu A, Mohlke KL, Nielsen JB, Okada Y, Peters U, Polderman TJC, Posthuma D, Reiner AP, Rice JP, Rimm E, Rose RJ, Runarsdottir V, Stallings MC, Stančáková A, Stefansson H, Thai KK, Tindle HA, Tyrfingsson T, Wall TL, Weir DR, Weisner C, Whitfield JB, Winsvold BS, Yin J, Zuccolo L, Bierut LJ, Hveem K, Lee JJ, Munafò MR, Saccone NL, Willer CJ, Cornelis MC, David SP, Hinds DA, Jorgenson E, Kaprio J, Stitzel JA, Stefansson K, Thorgeirsson TE, Abecasis G, Liu DJ, Vrieze S. Association studies of up to 1.2 million individuals yield new insights into the genetic etiology of tobacco and alcohol use. Nat Genet 2019; 51:237-244. [PMID: 30643251 PMCID: PMC6358542 DOI: 10.1038/s41588-018-0307-5] [Show More Authors] [Citation(s) in RCA: 1275] [Impact Index Per Article: 212.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 11/06/2018] [Indexed: 12/21/2022]
Abstract
Tobacco and alcohol use are leading causes of mortality that influence risk for many complex diseases and disorders1. They are heritable2,3 and etiologically related4,5 behaviors that have been resistant to gene discovery efforts6-11. In sample sizes up to 1.2 million individuals, we discovered 566 genetic variants in 406 loci associated with multiple stages of tobacco use (initiation, cessation, and heaviness) as well as alcohol use, with 150 loci evidencing pleiotropic association. Smoking phenotypes were positively genetically correlated with many health conditions, whereas alcohol use was negatively correlated with these conditions, such that increased genetic risk for alcohol use is associated with lower disease risk. We report evidence for the involvement of many systems in tobacco and alcohol use, including genes involved in nicotinic, dopaminergic, and glutamatergic neurotransmission. The results provide a solid starting point to evaluate the effects of these loci in model organisms and more precise substance use measures.
Collapse
Affiliation(s)
- Mengzhen Liu
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Yu Jiang
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, USA
- Institute of Personalized Medicine, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Robbee Wedow
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Sociology, University of Colorado Boulder, Boulder, CO, USA
- Institute of Behavioral Science, University of Colorado Boulder, Boulder, CO, USA
| | - Yue Li
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David M Brazel
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
- Interdisciplinary Quantitative Biology Graduate Group, University of Colorado Boulder, Boulder, CO, USA
| | - Fang Chen
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, USA
- Institute of Personalized Medicine, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Gargi Datta
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Jose Davila-Velderrain
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel McGuire
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, USA
- Institute of Personalized Medicine, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Chao Tian
- 23andMe, Inc., Mountain View, CA, USA
| | - Xiaowei Zhan
- Quantitative Biomedical Research Center, Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for the Genetics of Host Defense, Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hélène Choquet
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Anna R Docherty
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry and Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Johanna R Foerster
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Lars G Fritsche
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Maiken Elvestad Gabrielsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Scott D Gordon
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Hongyan Huang
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Seon-Kyeong Jang
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Philip R Jansen
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Child and Adolescent Psychiatry, Erasmus MC Rotterdam, Rotterdam, the Netherlands
| | - Yueh Ling
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Nana Matoba
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - George McMahon
- Department of Population Health Science, Bristol Medical School, Oakfield Grove, Bristol, UK
| | - Antonella Mulas
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | - Valeria Orrù
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Anita Pandit
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | | | - Anne Heidi Skogholt
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jennifer A Smith
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Amy E Taylor
- Department of Population Health Science, Bristol Medical School, Oakfield Grove, Bristol, UK
| | - Constance Turman
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Hannah Young
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Kendra A Young
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Gregory J M Zajac
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | | | - Jason D Boardman
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Sociology, University of Colorado Boulder, Boulder, CO, USA
- Institute of Behavioral Science, University of Colorado Boulder, Boulder, CO, USA
| | - Michael Boehnke
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Chu Chen
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | | | - Charles B Eaton
- Department of Family Medicine and Community Health, Alpert Medical School, Brown University, Providence, RI, USA
| | - Marissa A Ehringer
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Tõnu Esko
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Edoardo Fiorillo
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Italy
| | - Nathan A Gillespie
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Daniel F Gudbjartsson
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Toomas Haller
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Kathleen Mullan Harris
- Department of Sociology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew C Heath
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - John K Hewitt
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Ian B Hickie
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
| | - John E Hokanson
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christian J Hopfer
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David J Hunter
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - William G Iacono
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Eric O Johnson
- Fellows Program, RTI International, Research Triangle Park, NC, USA
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew C Keller
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter Kraft
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kenneth S Krauter
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Markku Laakso
- Department of Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Penelope A Lind
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Anu Loukola
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Sharon M Lutz
- Department of Biostatistics and Bioinformatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Pamela A F Madden
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Nicholas G Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Matt McGue
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Matthew B McQueen
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Sarah E Medland
- Psychiatric Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jonas B Nielsen
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Yukinori Okada
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Tinca J C Polderman
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Clinical Genetics, VU Medical Centre Amsterdam, Amsterdam, the Netherlands
| | - Alexander P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - John P Rice
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Eric Rimm
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Richard J Rose
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | | | - Michael C Stallings
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Alena Stančáková
- Department of Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | | | - Khanh K Thai
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Hilary A Tindle
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | | | - Tamara L Wall
- Department of Psychiatry, University of California, San Diego, San Diego, CA, USA
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Constance Weisner
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - John B Whitfield
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Jie Yin
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Luisa Zuccolo
- Department of Population Health Science, Bristol Medical School, Oakfield Grove, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Laura J Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway
- Department of Medicine, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - James J Lee
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Marcus R Munafò
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- UK Centre for Tobacco and Alcohol Studies, School of Psychological Science, University of Bristol, Bristol, UK
| | - Nancy L Saccone
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Cristen J Willer
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Marilyn C Cornelis
- Department of Preventative Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sean P David
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Eric Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Kari Stefansson
- deCODE Genetics/Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | - Gonçalo Abecasis
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Dajiang J Liu
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, USA.
- Institute of Personalized Medicine, College of Medicine, Pennsylvania State University, Hershey, PA, USA.
| | - Scott Vrieze
- Department of Psychology, University of Minnesota Twin Cities, Minneapolis, MN, USA.
| |
Collapse
|
47
|
Sharp BM, Chen H. Neurogenetic determinants and mechanisms of addiction to nicotine and smoked tobacco. Eur J Neurosci 2018; 50:2164-2179. [DOI: 10.1111/ejn.14171] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/31/2018] [Accepted: 09/18/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Burt M. Sharp
- Department of Genetics, Genomics and Informatics College of Medicine University of Tennessee Health Science Center 19 S. Manassas, CRB #220 Memphis TN 38103 USA
| | - Hao Chen
- Department of Pharmacology University of Tennessee Health Science Center Memphis TN USA
| |
Collapse
|
48
|
Bozinoff N, Le Foll B. Understanding the implications of the biobehavioral basis of nicotine addiction and its impact on the efficacy of treatment. Expert Rev Respir Med 2018; 12:793-804. [PMID: 30092681 DOI: 10.1080/17476348.2018.1507736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Tobacco use remains the leading cause of preventable death in the United States. There are efficacious behavioral and pharmacological options for smoking cessation including three FDA approved therapies - nicotine replacement therapy, varenicline and bupropion. Nevertheless, uptake of smoking cessation treatments continues to be poor and there is a need for novel smoking cessation treatments. Areas covered: This article reviews the biobehavioral basis of nicotine addiction, its implications for smoking cessation treatments, the various neurotransmitter systems involved in nicotine addictive effects, and their potential therapeutic value. Included are discussions around the role of genetic factors in predicting response to pharmacotherapy and what we know about appropriate application of pharmacotherapy and behavioral interventions for tobacco use disorder. The evidence for harm reduction measures in individuals who are not willing or able to quit smoking is also reviewed. Expert commentary: Many neurotransmitter system targets have been investigated as a result of our understanding of the underlying neurobiology of tobacco use disorder, and there remain important targets that have yet to be fully explored. rTMS or combination therapies are proposed as possible novel strategies to improve smoking cessation.
Collapse
Affiliation(s)
- Nikki Bozinoff
- a Department of Family and Community Medicine , University of Toronto , Toronto , Canada.,b Addiction Medicine Service, Acute Care Program , Centre for Addiction and Mental Health , Toronto , Canada
| | - Bernard Le Foll
- a Department of Family and Community Medicine , University of Toronto , Toronto , Canada.,b Addiction Medicine Service, Acute Care Program , Centre for Addiction and Mental Health , Toronto , Canada.,c Department of Pharmacology and Toxicology, Psychiatry, Institute of Medical Sciences , University of Toronto , Toronto , Canada.,d Centre for Addiction and Mental Health , Campbell Family Mental Health Research Institute, CAMH , Toronto , Canada.,e Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health , Toronto , Canada
| |
Collapse
|
49
|
Ji X, Bossé Y, Landi MT, Gui J, Xiao X, Qian D, Joubert P, Lamontagne M, Li Y, Gorlov I, de Biasi M, Han Y, Gorlova O, Hung RJ, Wu X, McKay J, Zong X, Carreras-Torres R, Christiani DC, Caporaso N, Johansson M, Liu G, Bojesen SE, Le Marchand L, Albanes D, Bickeböller H, Aldrich MC, Bush WS, Tardon A, Rennert G, Chen C, Teare MD, Field JK, Kiemeney LA, Lazarus P, Haugen A, Lam S, Schabath MB, Andrew AS, Shen H, Hong YC, Yuan JM, Bertazzi PA, Pesatori AC, Ye Y, Diao N, Su L, Zhang R, Brhane Y, Leighl N, Johansen JS, Mellemgaard A, Saliba W, Haiman C, Wilkens L, Fernandez-Somoano A, Fernandez-Tardon G, van der Heijden EHFM, Kim JH, Dai J, Hu Z, Davies MPA, Marcus MW, Brunnström H, Manjer J, Melander O, Muller DC, Overvad K, Trichopoulou A, Tumino R, Doherty J, Goodman GE, Cox A, Taylor F, Woll P, Brüske I, Manz J, Muley T, Risch A, Rosenberger A, Grankvist K, Johansson M, Shepherd F, Tsao MS, Arnold SM, Haura EB, Bolca C, Holcatova I, Janout V, Kontic M, Lissowska J, Mukeria A, Ognjanovic S, Orlowski TM, Scelo G, Swiatkowska B, Zaridze D, Bakke P, Skaug V, Zienolddiny S, et alJi X, Bossé Y, Landi MT, Gui J, Xiao X, Qian D, Joubert P, Lamontagne M, Li Y, Gorlov I, de Biasi M, Han Y, Gorlova O, Hung RJ, Wu X, McKay J, Zong X, Carreras-Torres R, Christiani DC, Caporaso N, Johansson M, Liu G, Bojesen SE, Le Marchand L, Albanes D, Bickeböller H, Aldrich MC, Bush WS, Tardon A, Rennert G, Chen C, Teare MD, Field JK, Kiemeney LA, Lazarus P, Haugen A, Lam S, Schabath MB, Andrew AS, Shen H, Hong YC, Yuan JM, Bertazzi PA, Pesatori AC, Ye Y, Diao N, Su L, Zhang R, Brhane Y, Leighl N, Johansen JS, Mellemgaard A, Saliba W, Haiman C, Wilkens L, Fernandez-Somoano A, Fernandez-Tardon G, van der Heijden EHFM, Kim JH, Dai J, Hu Z, Davies MPA, Marcus MW, Brunnström H, Manjer J, Melander O, Muller DC, Overvad K, Trichopoulou A, Tumino R, Doherty J, Goodman GE, Cox A, Taylor F, Woll P, Brüske I, Manz J, Muley T, Risch A, Rosenberger A, Grankvist K, Johansson M, Shepherd F, Tsao MS, Arnold SM, Haura EB, Bolca C, Holcatova I, Janout V, Kontic M, Lissowska J, Mukeria A, Ognjanovic S, Orlowski TM, Scelo G, Swiatkowska B, Zaridze D, Bakke P, Skaug V, Zienolddiny S, Duell EJ, Butler LM, Koh WP, Gao YT, Houlston R, McLaughlin J, Stevens V, Nickle DC, Obeidat M, Timens W, Zhu B, Song L, Artigas MS, Tobin MD, Wain LV, Gu F, Byun J, Kamal A, Zhu D, Tyndale RF, Wei WQ, Chanock S, Brennan P, Amos CI. Identification of susceptibility pathways for the role of chromosome 15q25.1 in modifying lung cancer risk. Nat Commun 2018; 9:3221. [PMID: 30104567 PMCID: PMC6089967 DOI: 10.1038/s41467-018-05074-y] [Show More Authors] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 05/01/2018] [Indexed: 12/20/2022] Open
Abstract
Genome-wide association studies (GWAS) identified the chromosome 15q25.1 locus as a leading susceptibility region for lung cancer. However, the pathogenic pathways, through which susceptibility SNPs within chromosome 15q25.1 affects lung cancer risk, have not been explored. We analyzed three cohorts with GWAS data consisting 42,901 individuals and lung expression quantitative trait loci (eQTL) data on 409 individuals to identify and validate the underlying pathways and to investigate the combined effect of genes from the identified susceptibility pathways. The KEGG neuroactive ligand receptor interaction pathway, two Reactome pathways, and 22 Gene Ontology terms were identified and replicated to be significantly associated with lung cancer risk, with P values less than 0.05 and FDR less than 0.1. Functional annotation of eQTL analysis results showed that the neuroactive ligand receptor interaction pathway and gated channel activity were involved in lung cancer risk. These pathways provide important insights for the etiology of lung cancer.
Collapse
Grants
- P30 CA023108 NCI NIH HHS
- P30 CA076292 NCI NIH HHS
- U01 CA063464 NCI NIH HHS
- P50 CA070907 NCI NIH HHS
- R01 CA111703 NCI NIH HHS
- UM1 CA182876 NCI NIH HHS
- UL1 TR000117 NCATS NIH HHS
- P20 CA090578 NCI NIH HHS
- U19 CA148127 NCI NIH HHS
- P20 GM103534 NIGMS NIH HHS
- UL1 TR000445 NCATS NIH HHS
- R01 LM012012 NLM NIH HHS
- R01 CA092824 NCI NIH HHS
- R35 CA197449 NCI NIH HHS
- UM1 CA164973 NCI NIH HHS
- U01 CA167462 NCI NIH HHS
- U19 CA203654 NCI NIH HHS
- R01 CA144034 NCI NIH HHS
- P20 RR018787 NCRR NIH HHS
- S10 RR025141 NCRR NIH HHS
- R01 CA074386 NCI NIH HHS
- R01 CA176568 NCI NIH HHS
- K07 CA172294 NCI NIH HHS
- P50 CA119997 NCI NIH HHS
- G0902313 Medical Research Council
- R01 CA063464 NCI NIH HHS
- P01 CA033619 NCI NIH HHS
- R01 HL133786 NHLBI NIH HHS
- P30 CA177558 NCI NIH HHS
- P50 CA090578 NCI NIH HHS
- U01 HG004798 NHGRI NIH HHS
- R01 CA151989 NCI NIH HHS
- 001 World Health Organization
- 202849/Z/16/Z Wellcome Trust
- UM1 CA167462 NCI NIH HHS
- U01 CA164973 NCI NIH HHS
- This work was supported by National Institutes of Health (NIH) for the research of lung cancer (grant P30CA023108, P20GM103534 and R01LM012012); Trandisciplinary Research in Cancer of the Lung (TRICL) (grant U19CA148127); UICC American Cancer Society Beginning Investigators Fellowship funded by the Union for International Cancer Control (UICC) (to X.Ji). CAPUA study. This work was supported by FIS-FEDER/Spain grant numbers FIS-01/310, FIS-PI03-0365, and FIS-07-BI060604, FICYT/Asturias grant numbers FICYT PB02-67 and FICYT IB09-133, and the University Institute of Oncology (IUOPA), of the University of Oviedo and the Ciber de Epidemiologia y Salud Pública. CIBERESP, SPAIN. The work performed in the CARET study was supported by the The National Institute of Health / National Cancer Institute: UM1 CA167462 (PI: Goodman), National Institute of Health UO1-CA6367307 (PIs Omen, Goodman); National Institute of Health R01 CA111703 (PI Chen), National Institute of Health 5R01 CA151989-01A1(PI Doherty). The Liverpool Lung project is supported by the Roy Castle Lung Cancer Foundation. The Harvard Lung Cancer Study was supported by the NIH (National Cancer Institute) grants CA092824, CA090578, CA074386 The Multiethnic Cohort Study was partially supported by NIH Grants CA164973, CA033619, CA63464 and CA148127 The work performed in MSH-PMH study was supported by The Canadian Cancer Society Research Institute (020214), Ontario Institute of Cancer and Cancer Care Ontario Chair Award to R.J.H. and G.L. and the Alan Brown Chair and Lusi Wong Programs at the Princess Margaret Hospital Foundation. NJLCS was funded by the State Key Program of National Natural Science of China (81230067), the National Key Basic Research Program Grant (2011CB503805), the Major Program of the National Natural Science Foundation of China (81390543). Norway study was supported by Norwegian Cancer Society, Norwegian Research Council The Shanghai Cohort Study (SCS) was supported by National Institutes of Health R01 CA144034 (PI: Yuan) and UM1 CA182876 (PI: Yuan). The Singapore Chinese Health Study (SCHS) was supported by National Institutes of Health R01 CA144034 (PI: Yuan) and UM1 CA182876 (PI: Yuan). The work in TLC study has been supported in part the James & Esther King Biomedical Research Program (09KN-15), National Institutes of Health Specialized Programs of Research Excellence (SPORE) Grant (P50 CA119997), and by a Cancer Center Support Grant (CCSG) at the H. Lee Moffitt Cancer Center and Research Institute, an NCI designated Comprehensive Cancer Center (grant number P30-CA76292) The Vanderbilt Lung Cancer Study – BioVU dataset used for the analyses described was obtained from Vanderbilt University Medical Center’s BioVU, which is supported by institutional funding, the 1S10RR025141-01 instrumentation award, and by the Vanderbilt CTSA grant UL1TR000445 from NCATS/NIH. Dr. Aldrich was supported by NIH/National Cancer Institute K07CA172294 (PI: Aldrich) and Dr. Bush was supported by NHGRI/NIH U01HG004798 (PI: Crawford). The Copenhagen General Population Study (CGPS) was supported by the Chief Physician Johan Boserup and Lise Boserup Fund, the Danish Medical Research Council and Herlev Hospital. The NELCS study: Grant Number P20RR018787 from the National Center for Research Resources (NCRR), a component of the National Institutes of Health (NIH). The MDACC study was supported in part by grants from the NIH (P50 CA070907, R01 CA176568) (to X. Wu), Cancer Prevention & Research Institute of Texas (RP130502) (to X. Wu), and The University of Texas MD Anderson Cancer Center institutional support for the Center for Translational and Public Health Genomics. The study in Lodz center was partially funded by Nofer Institute of Occupational Medicine, under task NIOM 10.13: Predictors of mortality from non-small cell lung cancer - field study. Kentucky Lung Cancer Research Initiative was supported by the Department of Defense [Congressionally Directed Medical Research Program, U.S. Army Medical Research and Materiel Command Program] under award number: 10153006 (W81XWH-11-1-0781). Views and opinions of, and endorsements by the author(s) do not reflect those of the US Army or the Department of Defense. This research was also supported by unrestricted infrastructure funds from the UK Center for Clinical and Translational Science, NIH grant UL1TR000117 and Markey Cancer Center NCI Cancer Center Support Grant (P30 CA177558) Shared Resource Facilities: Cancer Research Informatics, Biospecimen and Tissue Procurement, and Biostatistics and Bioinformatics. The Resource for the Study of Lung Cancer Epidemiology in North Trent (ReSoLuCENT) study was funded by the Sheffield Hospitals Charity, Sheffield Experimental Cancer Medicine Centre and Weston Park Hospital Cancer Charity. FT was supported by a clinical PhD fellowship funded by the Yorkshire Cancer Research/Cancer Research UK Sheffield Cancer Centre. The authors would like to thank the staff at the Respiratory Health Network Tissue Bank of the FRQS for their valuable assistance with the lung eQTL dataset at Laval University. The lung eQTL study at Laval University was supported by the Fondation de l’Institut universitaire de cardiologie et de pneumologie de Québec, the Respiratory Health Network of the FRQS, the Canadian Institutes of Health Research (MOP - 123369). Y.B. holds a Canada Research Chair in Genomics of Heart and Lung Diseases. The research undertaken by M.D.T., L.V.W. and M.S.A. was partly funded by the National Institute for Health Research (NIHR). The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR or the Department of Health. M.D.T. holds a Medical Research Council Senior Clinical Fellowship (G0902313).
Collapse
Affiliation(s)
- Xuemei Ji
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03750, NH, USA
| | - Yohan Bossé
- Department of Molecular Medicine, Laval University, Québec, G1V 4G5, Canada
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, G1V 4G5, Canada
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Jiang Gui
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03750, NH, USA
| | - Xiangjun Xiao
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03750, NH, USA
| | - David Qian
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03750, NH, USA
| | - Philippe Joubert
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, G1V 4G5, Canada
| | - Maxime Lamontagne
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, G1V 4G5, Canada
| | - Yafang Li
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03750, NH, USA
| | - Ivan Gorlov
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03750, NH, USA
| | - Mariella de Biasi
- Annenberg School of Communication, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Younghun Han
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03750, NH, USA
| | - Olga Gorlova
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03750, NH, USA
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, M5T 3L9, Canada
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, 77030, TX, USA
| | - James McKay
- International Agency for Research on Cancer, World Health Organization, Lyon, 69372 CEDEX 08, France
| | - Xuchen Zong
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, M5T 3L9, Canada
| | - Robert Carreras-Torres
- International Agency for Research on Cancer, World Health Organization, Lyon, 69372 CEDEX 08, France
| | - David C Christiani
- Department of Environmental Health, Harvard School of Public Health, Boston, 02115, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, 02115, MA, USA
| | - Neil Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Mattias Johansson
- International Agency for Research on Cancer, World Health Organization, Lyon, 69372 CEDEX 08, France
| | - Geoffrey Liu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, M5T 3L9, Canada
| | - Stig E Bojesen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Herlev 2730, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200 København N, Denmark
- Copenhagen General Population Study, Herlev and Gentofte Hospital, Ringvej 75, Copenhagen, Herlev 2730, Denmark
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, 96813, HI, USA
| | - Demetrios Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Heike Bickeböller
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, 37073, Germany
| | - Melinda C Aldrich
- Department of Thoracic Surgery, Division of Epidemiology, Vanderbilt University Medical Center, Nashville, 37203, TN, USA
| | - William S Bush
- Department of Thoracic Surgery, Division of Epidemiology, Vanderbilt University Medical Center, Nashville, 37203, TN, USA
- Department of Epidemiology and Biostatistics, School of Medicine, Case Western Reserve University, Cleveland, 44106, OH, USA
| | - Adonina Tardon
- Faculty of Medicine, University of Oviedo, Oviedo, 33006, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública, Campus del Cristo s/n, Oviedo, 33006, Spain
| | - Gad Rennert
- Clalit National Cancer Control Center, Carmel Medical Center, Haifa, 34361, Israel
- Faculty of Medicine, Technion, Haifa, 34361, Israel
| | - Chu Chen
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
| | - M Dawn Teare
- School of Health and Related Research, University of Sheffield, Sheffield, S1 4DA, UK
| | - John K Field
- Roy Castle Lung Cancer Research Programme, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - Lambertus A Kiemeney
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, 6525 EZ, The Netherlands
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, 99210-1495, WA, USA
| | - Aage Haugen
- National Institute of Occupational Health, 0033, Gydas vei 8, 0033, Oslo, Norway
| | - Stephen Lam
- British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, V5Z1L3, Canada
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, 33612, FL, USA
| | - Angeline S Andrew
- Department of Epidemiology, Geisel School of Medicine, 1 Medical Center Drive, Hanover, 03755, NH, USA
| | - Hongbing Shen
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, 101 Longmian Ave, Nanjing, 211166, PR China
| | - Yun-Chul Hong
- Department of Preventive Medicine, Seoul National University College of Medicine, 1 Gwanak-ro, Gwanak-gu, Seoul, 151 742, Republic of Korea
| | - Jian-Min Yuan
- University of Pittsburgh Cancer Institute, Pittsburgh, 15232, PA, USA
| | - Pier A Bertazzi
- Department of Preventive Medicine, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, Milan, 20133, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, 20133, Italy
| | - Angela C Pesatori
- Department of Preventive Medicine, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico, Milan, 20133, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, 20133, Italy
| | - Yuanqing Ye
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, 77030, TX, USA
| | - Nancy Diao
- Department of Environmental Health, Harvard School of Public Health, Boston, 02115, MA, USA
| | - Li Su
- Department of Environmental Health, Harvard School of Public Health, Boston, 02115, MA, USA
| | - Ruyang Zhang
- Department of Environmental Health, Harvard School of Public Health, Boston, 02115, MA, USA
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, 101 Longmian Ave, Nanjing, 211166, PR China
| | - Yonathan Brhane
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System and University of Toronto, Toronto, M5T 3L9, Canada
| | - Natasha Leighl
- University Health Network-The Princess Margaret Cancer Centre, 600 University Avenue, Toronto, M5G 2C4, Canada
| | - Jakob S Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, 2730, Denmark
| | - Anders Mellemgaard
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, 2730, Denmark
| | - Walid Saliba
- Clalit National Cancer Control Center, Carmel Medical Center, Haifa, 34361, Israel
- Faculty of Medicine, Technion, Haifa, 34361, Israel
| | - Christopher Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, 90033, CA, USA
| | - Lynne Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, 96813, HI, USA
| | - Ana Fernandez-Somoano
- Faculty of Medicine, University of Oviedo, Oviedo, 33006, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública, Campus del Cristo s/n, Oviedo, 33006, Spain
| | - Guillermo Fernandez-Tardon
- Faculty of Medicine, University of Oviedo, Oviedo, 33006, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública, Campus del Cristo s/n, Oviedo, 33006, Spain
| | - Erik H F M van der Heijden
- Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, 6525 EZ, The Netherlands
| | - Jin Hee Kim
- Department of Integrative Bioscience & Biotechnology, Sejong University, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, 101 Longmian Ave, Nanjing, 211166, PR China
| | - Zhibin Hu
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, 101 Longmian Ave, Nanjing, 211166, PR China
| | - Michael P A Davies
- Roy Castle Lung Cancer Research Programme, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - Michael W Marcus
- Roy Castle Lung Cancer Research Programme, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - Hans Brunnström
- Department of Pathology, Lund University, Lund, 222 41, Sweden
| | - Jonas Manjer
- Faculty of Medicine, Lund University, Lund, 22100, Sweden
| | - Olle Melander
- Faculty of Medicine, Lund University, Lund, 22100, Sweden
| | - David C Muller
- School of Public Health, St Mary's Campus, Imperial College London, London, W2 1PG, UK
| | - Kim Overvad
- Faculty of Medicine, Lund University, Lund, 22100, Sweden
| | | | - Rosario Tumino
- Cancer Registry and Histopathology Department, "Civic-M.P. Arezzo" Hospital, ASP, Ragusa, 97100, Italy
| | - Jennifer Doherty
- Department of Epidemiology, Geisel School of Medicine, 1 Medical Center Drive, Hanover, 03755, NH, USA
- Fred Hutchinson Cancer Research Center, Seattle, 98109-1024, WA, USA
| | - Gary E Goodman
- Fred Hutchinson Cancer Research Center, Seattle, 98109-1024, WA, USA
- Swedish Medical Group, Arnold Pavilion, Suite 200, Seattle, 98104, WA, USA
| | - Angela Cox
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2RX, UK
| | - Fiona Taylor
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2RX, UK
| | - Penella Woll
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2RX, UK
| | - Irene Brüske
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, D-85764, Germany
| | - Judith Manz
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, D-85764, Germany
| | - Thomas Muley
- Thoraxklinik at University Hospital Heidelberg, Heidelberg, 69126, Germany
- Translational Lung Research Center Heidelberg (TLRC-H), Heidelberg, 69120, Germany
| | - Angela Risch
- Cancer Cluster Salzburg, University of Salzburg, Salzburg, 5020, Austria
| | - Albert Rosenberger
- Department of Genetic Epidemiology, University Medical Center, Georg-August-University Göttingen, Göttingen, 37073, Germany
| | - Kjell Grankvist
- Department of Medical Biosciences, Umeå University, Umeå, 901 85, Sweden
| | - Mikael Johansson
- Department of Radiation Sciences, Umeå University, Umeå, 901 85, Sweden
| | | | | | - Susanne M Arnold
- Markey Cancer Center, University of Kentucky, First Floor, 800 Rose Street, Lexington, 40508, KY, USA
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, 33612, KY, USA
| | - Ciprian Bolca
- Institute of Pneumology "Marius Nasta", Bucharest, RO-050159, Romania
| | - Ivana Holcatova
- 1st Faculty of Medicine, Charles University, Kateřinská 32, Prague, 121 08 Praha 2, Czech Republic
| | - Vladimir Janout
- 1st Faculty of Medicine, Charles University, Kateřinská 32, Prague, 121 08 Praha 2, Czech Republic
| | - Milica Kontic
- Clinical Center of Serbia, Clinic for Pulmonology, School of Medicine, University of Belgrade, Belgrade, 11000, Serbia
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, M. Sklodowska-Curie Institute-Oncology Center, Warsaw, 02-781, Poland
| | - Anush Mukeria
- Department of Epidemiology and Prevention, Russian N.N. Blokhin Cancer Research Centre, Moscow, 115478, Russian Federation
| | - Simona Ognjanovic
- International Organization for Cancer Prevention and Research, Belgrade, 11070, Serbia
| | - Tadeusz M Orlowski
- Department of Surgery, National Tuberculosis and Lung Diseases Research Institute, Warsaw, PL-01-138, Poland
| | - Ghislaine Scelo
- International Agency for Research on Cancer, World Health Organization, Lyon, 69372 CEDEX 08, France
| | - Beata Swiatkowska
- Department of Environmental Epidemiology, Nofer Institute of Occupational Medicine, Lodz, 91-348, Poland
| | - David Zaridze
- Department of Epidemiology and Prevention, Russian N.N. Blokhin Cancer Research Centre, Moscow, 115478, Russian Federation
| | - Per Bakke
- Department of Clinical Science, University of Bergen, Bergen, 5021, Norway
| | - Vidar Skaug
- National Institute of Occupational Health, 0033, Gydas vei 8, 0033, Oslo, Norway
| | - Shanbeh Zienolddiny
- National Institute of Occupational Health, 0033, Gydas vei 8, 0033, Oslo, Norway
| | - Eric J Duell
- Unit of Nutrition and Cancer, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, 08908, Spain
| | - Lesley M Butler
- University of Pittsburgh Cancer Institute, Pittsburgh, 15232, PA, USA
| | - Woon-Puay Koh
- Duke-NUS Medical School, Singapore, 119077, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, 117549, Singapore
| | - Yu-Tang Gao
- Department of Epidemiology, Shanghai Cancer Institute, Shanghai, 2200, China
| | | | | | | | - David C Nickle
- Department of Genetics and Pharmacogenomics, Merck Research Laboratories, Boston, 02115-5727, MA, USA
| | - Ma'en Obeidat
- Centre for Heart Lung Innovation, St Paul's Hospital, The University of British Columbia, Vancouver, V6Z 1Y6, BC, Canada
| | - Wim Timens
- Department of Pathology and Medical Biology, GRIAC, University of Groningen, University Medical Center Groningen, Groningen, NL - 9713 GZ, The Netherlands
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Lei Song
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - María Soler Artigas
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
- Leicester Respiratory Biomedical Research Unit, National Institute for Health Research (NIHR), Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Martin D Tobin
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
- Leicester Respiratory Biomedical Research Unit, National Institute for Health Research (NIHR), Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Louise V Wain
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
- Leicester Respiratory Biomedical Research Unit, National Institute for Health Research (NIHR), Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Fangyi Gu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Jinyoung Byun
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03750, NH, USA
| | - Ahsan Kamal
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03750, NH, USA
| | - Dakai Zhu
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03750, NH, USA
| | - Rachel F Tyndale
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, M5T 1R8, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, M6J 1H4, ON, Canada
| | - Wei-Qi Wei
- Department of Biomedical Informatics, School of Medicine, Vanderbilt University, Nashville, TN, 37235, USA
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Paul Brennan
- International Agency for Research on Cancer, World Health Organization, Lyon, 69372 CEDEX 08, France
| | - Christopher I Amos
- Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, 03750, NH, USA.
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, 77030, TX, USA.
| |
Collapse
|
50
|
Morton G, Nasirova N, Sparks DW, Brodsky M, Sivakumaran S, Lambe EK, Turner EE. Chrna5-Expressing Neurons in the Interpeduncular Nucleus Mediate Aversion Primed by Prior Stimulation or Nicotine Exposure. J Neurosci 2018; 38:6900-6920. [PMID: 29954848 PMCID: PMC6070661 DOI: 10.1523/jneurosci.0023-18.2018] [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: 01/04/2018] [Revised: 06/08/2018] [Accepted: 06/15/2018] [Indexed: 11/21/2022] Open
Abstract
Genetic studies have shown an association between smoking and variation at the CHRNA5/A3/B4 gene locus encoding the α5, α3, and β4 nicotinic receptor subunits. The α5 receptor has been specifically implicated because smoking-associated haplotypes contain a coding variant in the CHRNA5 gene. The Chrna5/a3/b4 locus is conserved in rodents and the restricted expression of these subunits suggests neural pathways through which the reinforcing and aversive properties of nicotine may be mediated. Here, we show that, in the interpeduncular nucleus (IP), the site of the highest Chrna5 mRNA expression in rodents, electrophysiological responses to nicotinic acetylcholine receptor stimulation are markedly reduced in α5-null mice. IP neurons differ markedly from their upstream ventral medial habenula cholinergic partners, which appear unaltered by loss of α5. To probe the functional role of α5-containing IP neurons, we used BAC recombineering to generate transgenic mice expressing Cre-recombinase from the Chrna5 locus. Reporter expression driven by Chrna5Cre demonstrates that transcription of Chrna5 is regulated independently from the Chrna3/b4 genes transcribed on the opposite strand. Chrna5-expressing IP neurons are GABAergic and project to distant targets in the mesopontine raphe and tegmentum rather than forming local circuits. Optogenetic stimulation of Chrna5-expressing IP neurons failed to elicit physical manifestations of withdrawal. However, after recent prior stimulation or exposure to nicotine, IP stimulation becomes aversive. These results using mice of both sexes support the idea that the risk allele of CHRNA5 may increase the drive to smoke via loss of IP-mediated nicotine aversion.SIGNIFICANCE STATEMENT Understanding the receptors and neural pathways underlying the reinforcing and aversive effects of nicotine may suggest new treatments for tobacco addiction. Part of the individual variability in smoking is associated with specific forms of the α5 nicotinic receptor subunit gene. Here, we show that deletion of the α5 subunit in mice markedly reduces the cellular response to nicotine and acetylcholine in the interpeduncular nucleus (IP). Stimulation of α5-expressing IP neurons using optogenetics is aversive, but this effect requires priming by recent prior stimulation or exposure to nicotine. These results support the idea that the smoking-associated variant of the α5 gene may increase the drive to smoke via loss of IP-mediated nicotine aversion.
Collapse
Affiliation(s)
- Glenn Morton
- Center for Integrative Brain Research, Seattle Children's Research Institute
| | - Nailyam Nasirova
- Center for Integrative Brain Research, Seattle Children's Research Institute
| | | | - Matthew Brodsky
- Center for Integrative Brain Research, Seattle Children's Research Institute
| | | | - Evelyn K Lambe
- Department of Physiology
- Department of Obstetrics and Gynecology, and
- Department of Psychiatry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Eric E Turner
- Center for Integrative Brain Research, Seattle Children's Research Institute,
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington 98101
| |
Collapse
|