1
|
Hemminki K, Zitricky F, Sundquist K, Sundquist J, Försti A, Hemminki A. Sex specific familial risk in lung cancer through changing histologies in Sweden. Int J Cancer 2025. [PMID: 40156379 DOI: 10.1002/ijc.35431] [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] [Received: 11/14/2024] [Revised: 03/09/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
Familial clustering of lung cancer (LC) is related to shared smoking habits but the contribution of other potential factors such as sex or histology is not well known, and these are the subjects of the present study in Sweden. Family relationships (from Multigeneration register) and diagnosed cancers (from Cancer registry) were obtained from the national registers from 1961 to 2021. The overall familial risk for LC was constant from the 1990s but the male familial risk decreased while the female familial risk doubled at the same time when female incidence doubled. The female familial risk for mother-daughter pairs was higher (SIR = 2.2 [2.0-2.3], N = 716) than for father-son pairs (SIR = 1.6 [1.5-1.8], N = 962). The histology-specific familial risks for adenocarcinoma, squamous cell carcinoma, small cell and large cell carcinoma were highest for concordant histology but also present for discordant histology. The number of family members diagnosed with LC was a strong determinant of familial risk. The novel results showed that familial risk of LC depends on the background incidence of LC and is higher for women compared to men. We demonstrated further an increased familial risk for each of the four histological types of LC which was higher for concordant than discordant histologies but was even detected between discordant histologies suggesting that LC histology is not a genetic trait.
Collapse
Affiliation(s)
- Kari Hemminki
- Biomedical Center, Faculty of Medicine, Charles University Pilsen, Pilsen, Czech Republic
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frantisek Zitricky
- Biomedical Center, Faculty of Medicine, Charles University Pilsen, Pilsen, Czech Republic
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
- University Clinic Primary Care Skåne, Skåne, Sweden
- Department of Family and Community Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
- University Clinic Primary Care Skåne, Skåne, Sweden
- Department of Family and Community Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA
| | - Asta Försti
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
2
|
Williamson TJ, Park ER, Warner ET, Rasmussen AW, Ostroff JS. Quitting smoking after a cancer diagnosis is associated with reductions in stigma and anxiety: A longitudinal mediation analysis. STIGMA AND HEALTH 2025; 10:73-82. [PMID: 40026684 PMCID: PMC11867195 DOI: 10.1037/sah0000461] [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: 03/05/2025]
Abstract
Internalized cancer stigma is high among cancer patients who smoke, but it is unknown whether the experience of stigma changes after quitting smoking post-diagnosis. Using data from an RCT of tobacco treatment, we conducted a secondary data analysis and hypothesized that 1) cancer patients who quit smoking would report greater reductions in internalized cancer stigma, compared to patients who did not quit and that 2) greater reductions in stigma would significantly mediate the relationship between smoking abstinence and subsequent decreases in anxiety and depressive symptoms. Participants (n=303; 56.1% female) were adults recruited from two comprehensive cancer centers who had smoked in the past 30 days, spoke English or Spanish, and were being treated for a recent diagnosis of cancer. Participants completed questionnaires at baseline, 3-month follow-up, and 6-month follow-up, and biochemically verified smoking abstinence was determined by participants' salivary cotinine or carbon monoxide levels. Smoking abstinence at 3-month follow-up was significantly associated with reductions in cancer-related stigma from baseline to 3-month follow-up (b = -1.50, p < .001), controlling for sociodemographic and medical covariates. Additionally, reductions in stigma were associated with reductions in anxiety at 6-month follow-up (b = 0.28, p < .05), but not depressive symptoms. Reductions in stigma significantly mediated the relationship between smoking abstinence and decreased anxiety (indirect effect = -0.42, p < .05), but not depressive symptoms. Smoking cessation may be associated with reduction in internalized cancer stigma. Thus, in addition to benefits for medical outcomes, quitting smoking post-diagnosis may improve psychosocial well-being.
Collapse
Affiliation(s)
- Timothy J. Williamson
- Department of Psychological Science, Loyola Marymount University, Los Angeles, CA, USA
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elyse R. Park
- Health Promotion and Resiliency Intervention Research Program, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Erica T. Warner
- Mongan Institute, Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Autumn W. Rasmussen
- Health Promotion and Resiliency Intervention Research Program, Massachusetts General Hospital, Boston, MA, USA
| | - Jamie S. Ostroff
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
3
|
Jareebi MA. The Association Between Smoking Behavior and the Risk of Hypertension: Review of the Observational and Genetic Evidence. J Multidiscip Healthc 2024; 17:3265-3281. [PMID: 39006872 PMCID: PMC11246652 DOI: 10.2147/jmdh.s470589] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
Background Cigarette smoking is one of the world's largest avoidable risk factors for morbidity and mortality. Numerous studies have investigated the association between smoking and hypertension (HTN). Although observational data and cross-sectional research often exhibit a link between smoking and HTN, establishing causation remains challenging owing to potential confounding variables. Mendelian randomization (MR), a genetic epidemiological technique that employs genetic variants as instrumental variables, offers a more robust approach for evaluating causal links. This review aimed to explore both the observational and causal relationships between smoking and the risk of HTN. Methodology A comprehensive literature search across major electronic databases was conducted to identify relevant observational and Mendelian randomization (MR) studies on smoking and HTN risk. Various characteristics were included during study selection, such as study design, exposure assessment, and age range. Standardized processes were used for data retrieval and quality evaluation. Results Analysis of observational data revealed a paradoxical association between smoking and the risk of HTN, where a lower risk was observed among current smokers when compared to non-smokers. However, observational analysis also presented a dose-response effect with greater smoking intensity showed a modest linear increase in HTN risk, and older smoking initiation was associated with a slight increase in HTN risk (compared with younger). In contrast, MR-based causal estimates provide inconsistent evidence regarding the causal relationship between smoking behavior and HTN. Some MR analyses indicated a potential causal link between smoking and HTN; but this was not consistent. Conclusion Observational studies suggest a paradoxical association between smoking and HTN. However, MR studies do not provide sufficient evidence to establish a causal relationship. Regardless, lifestyle variables remain crucial for overall health. Healthcare professionals should regularly assess smoking status and provide counseling for quitting. Further research is needed to clarify the underlying processes, identify mediators, and evaluate the interventions.
Collapse
Affiliation(s)
- Mohammad A Jareebi
- Department of Family and Community Medicine, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
4
|
Milder CM, Howard SC, Ellis ED, Golden AP, Cohen SS, Mumma MT, Leggett RW, French B, Zablotska LB, Boice JD. Third mortality follow-up of the Mallinckrodt uranium processing workers, 1942-2019. Int J Radiat Biol 2024; 100:161-175. [PMID: 37819879 PMCID: PMC10843089 DOI: 10.1080/09553002.2023.2267640] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 08/22/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
INTRODUCTION Mallinckrodt Chemical Works was a uranium processing facility during the Manhattan Project from 1942 to 1966. Thousands of workers were exposed to low-dose-rates of ionizing radiation from external and internal sources. This third follow-up of 2514 White male employees updates cancer and noncancer mortality potentially associated with radiation and silica dust. MATERIALS AND METHODS Individual, annualized organ doses were estimated from film badge records (n monitored = 2514), occupational chest x-rays (n = 2514), uranium urinalysis (n = 1868), radium intake through radon breath measurements (n = 487), and radon ambient measurements (n = 1356). Silica dust exposure from pitchblende processing was estimated (n = 1317). Vital status and cause of death determination through 2019 relied upon the National Death Index and Social Security Administration Epidemiological Vital Status Service. The analysis included standardized mortality ratios (SMRs), Cox proportional hazards, and Poisson regression models. RESULTS Vital status was confirmed for 99.4% of workers (84.0% deceased). For a dose weighting factor of 1 for intakes of uranium, radium, and radon decay products, the mean and median lung doses were 65.6 and 29.9 mGy, respectively. SMRs indicated a difference in health outcomes between salaried and hourly workers, and more brain cancer deaths than expected [SMR: 1.79; 95% confidence interval (CI): 1.14, 2.70]. No association was seen between radiation and lung cancer [hazard ratio (HR) at 100 mGy: 0.93; 95%CI: 0.78, 1.11]. The relationship between radiation and kidney cancer observed in the previous follow-up was maintained (HR at 100 mGy: 2.07; 95%CI: 1.12, 3.79). Cardiovascular disease (CVD) also increased significantly with heart dose (HR at 100 mGy: 1.11; 95%CI: 1.02, 1.21). Exposures to dust ≥23.6 mg/m3-year were associated with nonmalignant kidney disease (NMKD) (HR: 3.02; 95%CI: 1.12, 8.16) and kidney cancer combined with NMKD (HR: 2.46; 95%CI: 1.04, 5.81), though without evidence of a dose-response per 100 mg/m3-year. CONCLUSIONS This third follow-up of Mallinckrodt uranium processors reinforced the results of the previous studies. There was an excess of brain cancers compared with the US population, although no radiation dose-response was detected. The association between radiation and kidney cancer remained, though potentially due to few cases at higher doses. The association between levels of silica dust ≥23.6 mg/m3-year and NMKD also remained. No association was observed between radiation and lung cancer. A positive dose-response was observed between radiation and CVD; however, this association may be confounded by smoking, which was unmeasured. Future work will pool these data with other uranium processing worker cohorts within the Million Person Study.
Collapse
Affiliation(s)
- Cato M. Milder
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute
| | | | | | | | - Sarah S. Cohen
- EpidStrategies, a Division of ToxStrategies, Inc., Katy, TX
| | | | | | - Benjamin French
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lydia B. Zablotska
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, CA, USA
| | - John D. Boice
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, USA
- National Council on Radiation Protection and Measurements (NCRP), Bethesda, MD, USA
| |
Collapse
|
5
|
Jain R, Xu W. Artificial Intelligence based wrapper for high dimensional feature selection. BMC Bioinformatics 2023; 24:392. [PMID: 37853338 PMCID: PMC10585895 DOI: 10.1186/s12859-023-05502-x] [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: 09/29/2022] [Accepted: 09/27/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Feature selection is important in high dimensional data analysis. The wrapper approach is one of the ways to perform feature selection, but it is computationally intensive as it builds and evaluates models of multiple subsets of features. The existing wrapper algorithm primarily focuses on shortening the path to find an optimal feature set. However, it underutilizes the capability of feature subset models, which impacts feature selection and its predictive performance. METHOD AND RESULTS This study proposes a novel Artificial Intelligence based Wrapper (AIWrap) algorithm that integrates Artificial Intelligence (AI) with the existing wrapper algorithm. The algorithm develops a Performance Prediction Model using AI which predicts the model performance of any feature set and allows the wrapper algorithm to evaluate the feature subset performance in a model without building the model. The algorithm can make the wrapper algorithm more relevant for high-dimensional data. We evaluate the performance of this algorithm using simulated studies and real research studies. AIWrap shows better or at par feature selection and model prediction performance than standard penalized feature selection algorithms and wrapper algorithms. CONCLUSION AIWrap approach provides an alternative algorithm to the existing algorithms for feature selection. The current study focuses on AIWrap application in continuous cross-sectional data. However, it could be applied to other datasets like longitudinal, categorical and time-to-event biological data.
Collapse
Affiliation(s)
- Rahi Jain
- Biostatistics Department, Princess Margaret Cancer Research Centre, Toronto, ON, Canada
| | - Wei Xu
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
6
|
Kumaraguru M, L L, Priyadharsini VJ, I MA, S R. Identification of Pathogenic Missense Mutations in the CHRNA5 Gene: A Computational Approach. Cureus 2023; 15:e47519. [PMID: 38021533 PMCID: PMC10663970 DOI: 10.7759/cureus.47519] [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: 07/31/2023] [Accepted: 10/22/2023] [Indexed: 12/01/2023] Open
Abstract
Aim The CHRNA5/A3/B4 gene locus is closely related to nicotine dependence and other smoking-related disorders. Coupling genetic and clinical studies of nicotine dependence and smoking behaviors may open new avenues for medication development. The aim of this study is to investigate the functional missense mutations in the CHRNA5 gene. Methodology The Ensembl database was used to gather data on missense mutations of the human CHRNA5 gene. Computational tools viz. SIFT (Sorting Intolerant From Tolerant), PolyPhen (Polymorphism Phenotyping), PROVEAN (Protein Variation Effect Analyzer), I-Mutant, and MutPred were used to uncover the pathogenic mutations in the gene under investigation. Results Among 161 missense variants reported inthe CHRNA5 gene, 94 variants were found to be highly pathogenic. Moreover, 20 were pathogenic and 4 were not pathogenic. Conclusion The computational analysis disclosed harmful mutations in the CHRNA5 gene which could be potentially associated with smoking-related traits.
Collapse
Affiliation(s)
- Mahalakshmi Kumaraguru
- Public Health Dentistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Leelavathi L
- Public Health Dentistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Vijayashree J Priyadharsini
- Clinical Genetics, Saveetha Dental College & Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Meignana Arumugham I
- Public Health Dentistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Rajeshkumar S
- Pharmacology, Saveetha Dental College & Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
7
|
Mirambeaux Villalona R. Influence of Genes in the Individualization of Smoking Cessation Pharmacological Treatment. Arch Bronconeumol 2023; 59:546-547. [PMID: 36803937 DOI: 10.1016/j.arbres.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023]
|
8
|
Doğan B, Ayar B, Pirim D. Investigation of putative roles of smoking-associated salivary microbiome alterations on carcinogenesis by integrative in silico analysis. Comput Biol Chem 2023; 102:107805. [PMID: 36587566 DOI: 10.1016/j.compbiolchem.2022.107805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/27/2022]
Abstract
Growing evidence suggests that cigarette smoking alters the salivary microbiome composition and affects the risk of various complex diseases including cancer. However, the potential role of the smoking-associated microbiome in cancer development remains unexplained. Here, the putative roles of smoking-related microbiome alterations in carcinogenesis were investigated by in silico analysis and suggested evidence can be further explored by experimental methodologies. The Disbiome database was used to extract smoking-associated microbial taxa in saliva and taxon set enrichment analysis (TSEA) was conducted to identify the gene sets associated with extracted microbial taxa. We further analyzed the expression profiles of identified genes by using RNA-sequencing data from TCGA and GTEx projects. Associations of the genes with smoking-related phenotypes in cancer datasets were analyzed to prioritize genes for their interplay between smoking-related microbiome and carcinogenesis. Thirty-eight microbial taxa associated with smoking were included in the TSEA and this revealed sixteen genes that were significantly associated with smoking-associated microbial taxa. All genes were found to be differentially expressed in at least one cancer dataset, yet the ELF3 and CTSH were the most common differentially expressed genes giving significant results for several cancer types. Moreover, C2CD3, CTSH, DSC3, ELF3, RHOT2, and WSB2 showed statistically significant associations with smoking-related phenotypes in cancer datasets. This study provides in silico evidence for the potential roles of the salivary microbiome on carcinogenesis. The results shed light on the importance of smoking cessation strategies for cancer management and interventions to stratify smokers for their risk of smoking-induced carcinogenesis.
Collapse
Affiliation(s)
- Berkcan Doğan
- Bursa Uludag University, Institute of Health Science, Department of Translational Medicine, 16059 Bursa, Turkey; Bursa Uludag University, Faculty of Medicine, Department of Medical Genetics, 16059 Bursa, Turkey
| | - Berna Ayar
- Bursa Uludag University, Department of Molecular Biology and Genetics, 16059 Bursa, Turkey; Istinye University, Institute of Health Science, Department of Molecular Oncology, 34010 Istanbul, Turkey
| | - Dilek Pirim
- Bursa Uludag University, Institute of Health Science, Department of Translational Medicine, 16059 Bursa, Turkey; Bursa Uludag University, Department of Molecular Biology and Genetics, 16059 Bursa, Turkey.
| |
Collapse
|
9
|
Chellappan S. Smoking Cessation after Cancer Diagnosis and Enhanced Therapy Response: Mechanisms and Significance. Curr Oncol 2022; 29:9956-9969. [PMID: 36547196 PMCID: PMC9776692 DOI: 10.3390/curroncol29120782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
The adverse effects of smoking on human health have been recognized for several decades, especially in the context of cancer. The ability of tobacco smoke components, including tobacco-specific carcinogens and additive compounds such as nicotine, to initiate or promote tumor growth have been described in hundreds of studies. These investigations have revealed the tumor-promoting activities of nicotine and other tobacco smoke components and have also recognized the ability of these agents to suppress the efficacy of cancer therapy; it is now clear that smoking can reduce the efficacy of most of the widely used therapeutic modalities, including immunotherapy, radiation therapy, and chemotherapy. Several studies examined if continued smoking after cancer diagnosis affected therapy response; it was found that while never smokers or non-smokers had the best response to therapy, those who quit smoking at the time of diagnosis had higher overall survival and reduced side-effects than those who continued to smoke. These studies also revealed the multiple mechanisms via which smoking enhances the growth and survival of tumors while suppressing therapy-induced cell death. In conclusion, smoking cessation during the course of cancer therapy markedly increases the chances of survival and the quality of life.
Collapse
Affiliation(s)
- Srikumar Chellappan
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
10
|
Howell MP, Jones CW, Herman CA, Mayne CV, Fernandez C, Theall KP, Esteves KC, Drury SS. Impact of prenatal tobacco smoking on infant telomere length trajectory and ADHD symptoms at 18 months: a longitudinal cohort study. BMC Med 2022; 20:153. [PMID: 35477473 PMCID: PMC9047258 DOI: 10.1186/s12916-022-02340-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 03/14/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Prenatal maternal tobacco smoking is a predictor of child attention-deficit/hyperactivity disorder (ADHD) and is associated with offspring telomere length (TL). In this study, we examine the relationship between maternal prenatal smoking, infant TL, and maternal report of early childhood symptoms of ADHD. METHODS One-hundred and eighty-one mother-infant dyads were followed prospectively for the infant's first 18 months of life. Prenatal smoking was assessed from maternal report and medical records. TL was measured from infant buccal swab DNA obtained across the first 18 months of life. ADHD symptoms were obtained from maternal report on the Child Behavior Check List. Multiple regression models tested the relation between prenatal smoking and both ADHD symptoms and infant TL. Additional analyses tested whether the change in infant TL influenced the relation between prenatal smoking and ADHD symptoms. RESULTS Sixteen percent of mothers reported prenatal smoking. Infant TL at 4, 12, and 18 months of age were correlated. Consistent with previous cross-sectional studies linking shorter offspring TL to maternal prenatal smoking, maternal prenatal smoking predicted greater telomere shortening from four to 18 months of infant age (β = - 5.797, 95% CI [-10.207, -1.386]; p = 0.010). Maternal depression was positively associated with both prenatal smoking (odds ratio (OR): 4.614, 95% CI [1.733, 12.282]; p = 0.002) and child ADHD symptoms (β = 4.713, 95% CI [2.073, 7.354]; p = 0.0006). To prevent confounding, analyses examined the relation between TL, ADHD symptoms, and prenatal smoking only in non-depressed mothers. In non-depressed mothers, infant TL attrition across the first 18 months moderated the relation between smoking and child ADHD. CONCLUSIONS The findings extend previous studies linking prenatal smoking to shorter infant TL by providing data demonstrating the effect on TL trajectory. The relation between prenatal smoking and early infant ADHD symptoms was moderated by the change in TL. The findings provide novel initial evidence suggesting that TL dynamics are one mechanistic pathway influencing the relation between maternal prenatal smoking and ADHD.
Collapse
Affiliation(s)
- Meghan P Howell
- Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue, #8526, New Orleans, Louisiana, 70112, USA
| | - Christopher W Jones
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cade A Herman
- Tufts University School of Medicine, Boston, MA, USA
| | - Celia V Mayne
- Department of Psychiatry and Behavioral Science, Tulane University School of Medicine, 1430 Tulane Avenue, #8526, New Orleans, Louisiana, 70112, USA
| | - Camilo Fernandez
- Department of Orthopedic Surgery, Tulane University School of Medicine, 1430 Tulane Avenue, #8526, New Orleans, Louisiana, 70112, USA.,Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Katherine P Theall
- Global Community Health and Behavioral Sciences, Tulane University School of Public Health and Tropical Medicine, 1430 Tulane Avenue, #8526, New Orleans, Louisiana, 70112, USA
| | - Kyle C Esteves
- Clinical Neuroscience Research Center, Tulane University School of Medicine, 1430 Tulane Avenue, #8526, New Orleans, Louisiana, 70112, USA
| | - Stacy S Drury
- Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue, #8526, New Orleans, Louisiana, 70112, USA. .,Department of Psychiatry and Behavioral Science, Tulane University School of Medicine, 1430 Tulane Avenue, #8526, New Orleans, Louisiana, 70112, USA.
| |
Collapse
|
11
|
Ambrose JA, Najafi A, Jain V, Muller JE, Ranka S, Barua RS. Reducing Tobacco-Related Disability in Chronic Smokers. Am J Med 2020; 133:908-915. [PMID: 32325048 DOI: 10.1016/j.amjmed.2020.03.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 02/02/2023]
Abstract
Tobacco consumption (predominantly cigarettes) is the leading preventable cause of mortality worldwide. Although the major focus of strategies to reduce mortality from tobacco must include prevention of future generations from initially gaining access, some smokers are unwilling or unable to quit. Can the higher risk chronic smoker be identified and can their risk be reduced? The risk of adverse events in cigarette smokers is influenced by the intensity and duration of cigarette smoking or secondhand exposure, associated conventional risk factors, environmental stressors, and certain genetic variants and epigenetic modifiers. Recent data suggest that inflammatory markers such as high-sensitivity C-reactive protein (hs CRP) and targeted imaging can identify some smokers at higher risk. As smoking is prothrombotic, aspirin initiation and expanded statin use might reduce cardiovascular risk in those who do not presently meet criteria for these therapies, but further study is required. Thus, although advocacy for smoking cessation should always be the primary approach, increased efforts are needed to identify and potentially treat those who are unable or unwilling to quit.
Collapse
Affiliation(s)
- John A Ambrose
- University of California, San Francisco, Fresno Medical Education Program, Fresno, Calif.
| | - Amir Najafi
- University of California, San Francisco, Fresno Medical Education Program, Fresno, Calif
| | - Vipul Jain
- University of California, San Francisco, Fresno Medical Education Program, Fresno, Calif
| | | | - Sagar Ranka
- University of Kansas Medical Center, Kansas City Veterans' Administration, Kansas City, Mo
| | - Rajat S Barua
- University of Kansas Medical Center, Kansas City Veterans' Administration, Kansas City, Mo
| |
Collapse
|
12
|
Lee C, Gao M, Ryff CD. Conscientiousness and Smoking: Do Cultural Context and Gender Matter? Front Psychol 2020; 11:1593. [PMID: 32733344 PMCID: PMC7358448 DOI: 10.3389/fpsyg.2020.01593] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/15/2020] [Indexed: 01/26/2023] Open
Abstract
Prior studies have found that conscientiousness has a protective effect against smoking, but evidence for this relationship mostly comes from Western contexts. In societies where smoking is pervasive and less stigmatized, the protective effect of conscientiousness on smoking may be less evident. Moreover, whether smoking is viewed as normal or deviant also may vary by gender norms attached to smoking. Using surveys of Midlife Development in the United States (MIDUS) and Japan (MIDJA), we examined patterns in the association between conscientiousness and smoking status (never, former, current) for men and women. We found that in the United States, where the social unacceptability of smoking has dramatically increased, there is an inverse association between conscientiousness and smoking status for both genders. In Japan, where the stigma attached to smoking operates for women but not men, the association between conscientiousness and smoking status varies by gender. For Japanese men, levels of conscientiousness do not differ across smoking statuses. For Japanese women, those who formerly smoked show lower levels of conscientiousness than those who never smoked and those who currently smoke. We interpret these findings in light of differing cultural and historical backgrounds of smoking for men and women.
Collapse
Affiliation(s)
- Chioun Lee
- Department of Sociology, University of California, Riverside, Riverside, CA, United States,*Correspondence: Chioun Lee,
| | - Manjing Gao
- Department of Sociology, University of California, Riverside, Riverside, CA, United States
| | - Carol D. Ryff
- Institute on Aging and Department of Psychology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
13
|
Shaikh F, Kjølllesdal MK, Carslake D, Stoltenberg C, Davey Smith G, Næss Ø. Birthweight in offspring and cardiovascular mortality in their parents, aunts and uncles: a family-based cohort study of 1.35 million births. Int J Epidemiol 2020; 49:205-215. [PMID: 31325357 PMCID: PMC7124506 DOI: 10.1093/ije/dyz156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2019] [Indexed: 11/26/2022] Open
Abstract
Background A link between suboptimal fetal growth and higher risk of cardiovascular disease (CVD) is well documented. It has been difficult to assess the contribution of environmental versus genetic factors to the association, as these factors are closely connected in nuclear families. We investigated the association between offspring birthweight and CVD mortality in parents, aunts and uncles, and examined whether these associations are explained by CVD risk factors. Methods We linked Norwegian data from the Medical Birth Registry, the Cause of Death Registry and cardiovascular surveys. A total of 1 353 956 births (1967–2012) were linked to parents and one maternal and one paternal aunt/uncle. Offspring birthweight and CVD mortality association among all relationships was assessed by hazard ratios (HR) from Cox regressions. The influence of CVD risk factors on the associations was examined in a subgroup. Results Offspring birthweight was inversely associated with CVD mortality among parents and aunts/uncles. HR of CVD mortality for one standard deviation (SD) increase in offspring birthweight was 0.72 (0.69–0.75) in mothers and 0.89 (0.86–0.92) in fathers. In aunts/uncles, the HRs were between 0.90 (0.86–0.95) and 0.93 (0.91–0.95). Adjustment for CVD risk factors in a subgroup attenuated all the associations. Conclusions Birthweight was associated with increased risk of CVD in parents and in aunts/uncles. These associations were largely explained by CVD risk factors. Our findings suggest that associations between offspring birthweight and CVD in adult relatives involve both behavioural variables (especially smoking) and shared genetics relating to established CVD risk factors.
Collapse
Affiliation(s)
- Fareeha Shaikh
- Institute of Health and Society, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - David Carslake
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Camilla Stoltenberg
- Norwegian Institute of Public Health, Oslo, Norway.,Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - George Davey Smith
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK.,Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Øyvind Næss
- Institute of Health and Society, Faculty of Medicine, University of Oslo, Oslo, Norway.,Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
14
|
Li M, Chen Y, Yao J, Lu S, Guan Y, Xu Y, Liu Q, Sun S, Mi Q, Mei J, Li X, Miao M, Zhao S, Zhu Z. Genome-Wide Association Study of Smoking Behavior Traits in a Chinese Han Population. Front Psychiatry 2020; 11:564239. [PMID: 33033484 PMCID: PMC7509597 DOI: 10.3389/fpsyt.2020.564239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/17/2020] [Indexed: 01/12/2023] Open
Abstract
Tobacco use is one of the leading causes of preventable disease worldwide. Genetic studies have elucidated numerous smoking-associated risk loci in American and European populations. However, genetic determinants for cigarette smoking in Chinese populations are under investigated. In this study, a whole-genome sequencing (WGS)-based genome-wide association study (GWAS) was performed in a Chinese Han population comprising 620 smokers and 564 nonsmokers. Thirteen single-nucleotide polymorphisms (SNPs) of the raftlin lipid linker 1 (RFTN1) gene achieved genome-wide significance levels (P < 5 x 10-8) for smoking initiation. The rs139753473 from RFTN1 and six other suggestively significant loci from CUB and sushi multiple domains 1 (CSMD1) gene were also associated with cigarettes per day (CPD) in an independent Chinese sample consisting of 1,329 subjects (805 smokers and 524 nonsmokers). When treating males separately, associations between smoking initiation and PCAT5/ANKRD30A, two genes involved in cancer development, were identified and replicated. Within RFTN1, two haplotypes (i.e., C-A-C-G and A-G-T-C) formed by rs796812630-rs796584733-rs796349027-rs879511366 and three haplotypes (i.e., T-T-C-C-C, T-T-A-T-T, and C-A-A-T-T) formed by rs879401109-rs879453873-rs75180423-rs541378415-rs796757175 were strongly associated with smoking initiation. In addition, we also revealed two haplotypes (i.e., C-A-G-G and T-C-T-T derived from rs4875371-rs4875372-rs17070935-rs11991366) in the CSMD1 gene showing a significant association with smoking initiation. Further bioinformatics functional assessment suggested that RFTN1 may participate in smoking behavior through modulating immune responses or interactions with the glucocorticoid receptor alpha and the androgen receptor. Together, our results may help understand the mechanisms underlying smoking behavior in the Chinese Han population.
Collapse
Affiliation(s)
- Meng Li
- Joint Institute of Tobacco and Health, Yunnan Academy of Tobacco Science, Kunming, China
| | - Ying Chen
- Joint Institute of Tobacco and Health, Yunnan Academy of Tobacco Science, Kunming, China
| | - Jianhua Yao
- Joint Institute of Tobacco and Health, Yunnan Academy of Tobacco Science, Kunming, China
| | - Sheming Lu
- Joint Institute of Tobacco and Health, Yunnan Academy of Tobacco Science, Kunming, China
| | - Ying Guan
- Joint Institute of Tobacco and Health, Yunnan Academy of Tobacco Science, Kunming, China
| | - Yuqiong Xu
- Joint Institute of Tobacco and Health, Yunnan Academy of Tobacco Science, Kunming, China
| | - Qiang Liu
- Hangzhou Global Biotechnology and Bioinformatics Co. Ltd, Hangzhou, China
| | - Silong Sun
- Joint Institute of Tobacco and Health, Yunnan Academy of Tobacco Science, Kunming, China
| | - Qili Mi
- Joint Institute of Tobacco and Health, Yunnan Academy of Tobacco Science, Kunming, China
| | - Junpu Mei
- Joint Institute of Tobacco and Health, Yunnan Academy of Tobacco Science, Kunming, China
| | - Xuemei Li
- Joint Institute of Tobacco and Health, Yunnan Academy of Tobacco Science, Kunming, China
| | - Mingming Miao
- Joint Institute of Tobacco and Health, Yunnan Academy of Tobacco Science, Kunming, China
| | - Shancen Zhao
- Joint Institute of Tobacco and Health, Yunnan Academy of Tobacco Science, Kunming, China
| | - Zhouhai Zhu
- Joint Institute of Tobacco and Health, Yunnan Academy of Tobacco Science, Kunming, China
| |
Collapse
|
15
|
Mørkve Knudsen GT, Rezwan FI, Johannessen A, Skulstad SM, Bertelsen RJ, Real FG, Krauss-Etschmann S, Patil V, Jarvis D, Arshad SH, Holloway JW, Svanes C. Epigenome-wide association of father's smoking with offspring DNA methylation: a hypothesis-generating study. ENVIRONMENTAL EPIGENETICS 2019; 5:dvz023. [PMID: 31827900 PMCID: PMC6896979 DOI: 10.1093/eep/dvz023] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 08/22/2019] [Accepted: 11/04/2019] [Indexed: 05/23/2023]
Abstract
Epidemiological studies suggest that father's smoking might influence their future children's health, but few studies have addressed whether paternal line effects might be related to altered DNA methylation patterns in the offspring. To investigate a potential association between fathers' smoking exposures and offspring DNA methylation using epigenome-wide association studies. We used data from 195 males and females (11-54 years) participating in two population-based cohorts. DNA methylation was quantified in whole blood using Illumina Infinium MethylationEPIC Beadchip. Comb-p was used to analyse differentially methylated regions (DMRs). Robust multivariate linear models, adjusted for personal/maternal smoking and cell-type proportion, were used to analyse offspring differentially associated probes (DMPs) related to paternal smoking. In sensitivity analyses, we adjusted for socio-economic position and clustering by family. Adjustment for inflation was based on estimation of the empirical null distribution in BACON. Enrichment and pathway analyses were performed on genes annotated to cytosine-phosphate-guanine (CpG) sites using the gometh function in missMethyl. We identified six significant DMRs (Sidak-corrected P values: 0.0006-0.0173), associated with paternal smoking, annotated to genes involved in innate and adaptive immunity, fatty acid synthesis, development and function of neuronal systems and cellular processes. DMP analysis identified 33 CpGs [false discovery rate (FDR) < 0.05]. Following adjustment for genomic control (λ = 1.462), no DMPs remained epigenome-wide significant (FDR < 0.05). This hypothesis-generating study found that fathers' smoking was associated with differential methylation in their adolescent and adult offspring. Future studies are needed to explore the intriguing hypothesis that fathers' exposures might persistently modify their future offspring's epigenome.
Collapse
Affiliation(s)
- G T Mørkve Knudsen
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
- Department of Occupational Medicine, Haukeland University Hospital, N-5021 Bergen, Norway
- Correspondence address. Haukanesvegen 260, N-5650 Tysse, Norway; Tel: +47 977 98 147; E-mail: and
| | - F I Rezwan
- Human Genetics and Genomic Medicine, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - A Johannessen
- Department of Occupational Medicine, Haukeland University Hospital, N-5021 Bergen, Norway
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, N-5018 Bergen, Norway
| | - S M Skulstad
- Department of Occupational Medicine, Haukeland University Hospital, N-5021 Bergen, Norway
| | - R J Bertelsen
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
| | - F G Real
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway
| | - S Krauss-Etschmann
- Division of Experimental Asthma Research, Research Center Borstel, 23845 Borstel, Germany
- German Center for Lung Research (DZL) and Institute of Experimental Medicine, Christian-Albrechts University of Kiel, 24118 Kiel, Germany
| | - V Patil
- David Hide Asthma and Allergy Research Centre, St. Mary’s Hospital, Isle of Wight PO30 5TG, UK
| | - D Jarvis
- Faculty of Medicine, National Heart & Lung Institute, Imperial College, London SW3 6LY, UK
| | - S H Arshad
- Clinical and Experimental Sciences, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
- NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton SO16 6YD, UK
| | - J W Holloway
- Human Genetics and Genomic Medicine, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - C Svanes
- Department of Occupational Medicine, Haukeland University Hospital, N-5021 Bergen, Norway
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, N-5018 Bergen, Norway
| |
Collapse
|
16
|
Mbarek H, van Beijsterveldt CEM, Jan Hottenga J, Dolan CV, Boomsma DI, Willemsen G, Vink JM. Association Between rs1051730 and Smoking During Pregnancy in Dutch Women. Nicotine Tob Res 2019; 21:835-840. [PMID: 29228387 PMCID: PMC6528154 DOI: 10.1093/ntr/ntx267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 12/05/2017] [Indexed: 01/09/2023]
Abstract
INTRODUCTION The common genetic variant (rs1051730) in the 15q24 nicotinic acetylcholine receptor gene cluster CHRNA5-CHRNA3-CHRNB4 was associated with smoking quantity and has been reported to be associated also with reduced ability to quit smoking in pregnant women but results were inconsistent in nonpregnant women. The aim of this study was to explore the association between rs1051730 and smoking cessation during pregnancy in a sample of Dutch women. METHODS Data on smoking during pregnancy were available from 1337 women, who ever smoked, registered at the Netherlands Twin Register (NTR). Logistic regression was used to assess evidence for the association of rs1051730 genotype on smoking during pregnancy. In a subsample of 561 women, we investigated the influence of partner's smoking. Educational attainment and year of birth were used as covariates in both analyses. RESULTS There was evidence for a significant association between having one or more T alleles of the rs1051730 polymorphism and the likelihood of smoking during pregnancy (p = .03, odds ratio = 1.28, 95% CI = 1.02 to 1.61). However, this association attenuated when adjusting for birth cohort and educational attainment (p = .37, odds ratio = 1.12, 95% CI = 0.87 to 1.43). In the subsample, smoking spouse was highly associated with smoking during pregnancy, even when educational attainment and birth cohort were included in the model. CONCLUSIONS Our results did not support a strong association between this genetic variant and smoking during pregnancy. However, a strong association was observed with the smoking behavior of the partner, regardless of the genotype of the women. IMPLICATIONS The present study emphasizes the importance of social influences like spousal smoking on the smoking behavior of pregnant women. Further research is needed to address the role of rs1051730 genetic variant in influencing smoking cessation and the interaction with important environmental factors like the smoking behavior of the partner.
Collapse
Affiliation(s)
- Hamdi Mbarek
- Department of Biological Psychology, Amsterdam Public Health research institute, Vrije Universiteit, Amsterdam, The Netherlands
| | - Catharina E M van Beijsterveldt
- Department of Biological Psychology, Amsterdam Public Health research institute, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jouke Jan Hottenga
- Department of Biological Psychology, Amsterdam Public Health research institute, Vrije Universiteit, Amsterdam, The Netherlands
| | - Conor V Dolan
- Department of Biological Psychology, Amsterdam Public Health research institute, Vrije Universiteit, Amsterdam, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Amsterdam Public Health research institute, Vrije Universiteit, Amsterdam, The Netherlands
| | - Gonneke Willemsen
- Department of Biological Psychology, Amsterdam Public Health research institute, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jacqueline M Vink
- Department of Biological Psychology, Amsterdam Public Health research institute, Vrije Universiteit, Amsterdam, The Netherlands
- Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
17
|
Garcia‐Rivas V, Deroche‐Gamonet V. Not all smokers appear to seek nicotine for the same reasons: implications for preclinical research in nicotine dependence. Addict Biol 2019; 24:317-334. [PMID: 29480575 DOI: 10.1111/adb.12607] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 12/11/2017] [Accepted: 01/15/2018] [Indexed: 01/08/2023]
Abstract
Tobacco use leads to 6 million deaths every year due to severe long-lasting diseases. The main component of tobacco, nicotine, is recognized as one of the most addictive drugs, making smoking cessation difficult, even when 70 percent of smokers wish to do so. Clinical and preclinical studies have demonstrated consistently that nicotine seeking is a complex behavior involving various psychopharmacological mechanisms. Evidence supports that the population of smokers is heterogeneous, particularly as regards the breadth of motives that determine the urge to smoke. Here, we review converging psychological, genetic and neurobiological data from clinical and preclinical studies supporting that the mechanisms controlling nicotine seeking may vary from individual to individual. It appears timely that basic neuroscience integrates this heterogeneity to refine our understanding of the neurobiology of nicotine seeking, as tremendous progress has been made in modeling the various psychopharmacological mechanisms driving nicotine seeking in rodents. For a better understanding of the mechanisms that drive nicotine seeking, we emphasize the need for individual-based research strategies in which nicotine seeking, and eventually treatment efficacy, are determined while taking into account individual variations in the mechanisms of nicotine seeking.
Collapse
Affiliation(s)
- Vernon Garcia‐Rivas
- Université de Bordeaux France
- INSERM U1215, Psychobiology of Drug AddictionNeuroCentre Magendie France
| | - Véronique Deroche‐Gamonet
- Université de Bordeaux France
- INSERM U1215, Psychobiology of Drug AddictionNeuroCentre Magendie France
| |
Collapse
|
18
|
Andersson C, Lukács Krogager M, Kuhr Skals R, Rosenbaum Appel EV, Theil Have C, Grarup N, Pedersen O, Jeppesen JL, Pedersen OD, Dominguez H, Dixen U, Engstrøm T, Tønder N, Roden DM, Stender S, Gislason GH, Enghusen-Poulsen H, Hansen T, Køber L, Torp-Pedersen C, Weeke PE. Association of genetic variants previously implicated in coronary artery disease with age at onset of coronary artery disease requiring revascularizations. PLoS One 2019; 14:e0211690. [PMID: 30726294 PMCID: PMC6364925 DOI: 10.1371/journal.pone.0211690] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 01/20/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The relation between burden of risk factors, familial coronary artery disease (CAD), and known genetic variants underlying CAD and low-density lipoprotein cholesterol (LDL-C) levels is not well-explored in clinical samples. We aimed to investigate the association of these measures with age at onset of CAD requiring revascularizations in a clinical sample of patients undergoing first-time coronary angiography. METHODS 1599 individuals (mean age 64 years [min-max 29-96 years], 28% women) were genotyped (from blood drawn as part of usual clinical care) in the Copenhagen area (2010-2014). The burden of common genetic variants was measured as aggregated genetic risk scores (GRS) of single nucleotide polymorphisms (SNPs) discovered in genome-wide association studies. RESULTS Self-reported familial CAD (prevalent in 41% of the sample) was associated with -3.2 years (95% confidence interval -4.5, -2.2, p<0.0001) earlier need of revascularization in sex-adjusted models. Patients with and without familial CAD had similar mean values of CAD-GRS (unweighted scores 68.4 vs. 68.0, p = 0.10, weighted scores 67.7 vs. 67.5, p = 0.49) and LDL-C-GRS (unweighted scores 58.5 vs. 58.3, p = 0.34, weighted scores 63.3 vs. 61.1, p = 0.41). The correlation between the CAD-GRS and LDL-C-GRS was low (r = 0.14, p<0.001). In multivariable adjusted regression models, each 1 standard deviation higher values of LDL-C-GRS and CAD-GRS were associated with -0.70 years (95% confidence interval -1.25, -0.14, p = 0.014) and -0.51 years (-1.07, 0.04, p = 0.07) earlier need for revascularization, respectively. CONCLUSIONS Young individuals presenting with CAD requiring surgical interventions had a higher genetic burden of SNPs relating to LDL-C and CAD (although the latter was statistically non-significant), compared with older individuals. However, the absolute difference was modest, suggesting that genetic screening can currently not be used as an effective prediction tool of when in life a person will develop CAD. Whether undiscovered genetic variants can still explain a "missing heritability" in early-onset CAD warrants more research.
Collapse
Affiliation(s)
- Charlotte Andersson
- Department of Cardiology, Herlev and Gentofte Hospital, Hellerup, Denmark
- * E-mail:
| | - Maria Lukács Krogager
- Unit of Epidemiology and biostatistics, Aalborg University Hospital, Aalborg, Denmark
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Regitze Kuhr Skals
- Unit of Epidemiology and biostatistics, Aalborg University Hospital, Aalborg, Denmark
| | - Emil Vincent Rosenbaum Appel
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian Theil Have
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen L. Jeppesen
- Department of Internal Medicine, Section of Cardiology, Amager and Hvidovre Hospital Glostrup, Glostrup, Denmark
| | | | - Helena Dominguez
- Department of Cardiology, Bispebjerg and Frederiksberg Hospital, Bispebjerg, Denmark
- Department of Biomedicine, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Dixen
- Department of Cardiology, Amager and Hvidovre Hospital Hvidovre, Hvidovre, Denmark
| | - Thomas Engstrøm
- Department of Cardiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Niels Tønder
- Department of Cardiology, Nephrology, and Endocrinology, Hillerød Hospital, Hillerød, Denmark
| | - Dan M. Roden
- Departments of Medicine, Pharmacology, and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Steen Stender
- Department of Clinical Biochemistry, Copenhagen University Hospital, Gentofte, Denmark
| | - Gunnar H. Gislason
- Department of Cardiology, Herlev and Gentofte Hospital, Hellerup, Denmark
- The Danish Heart Foundation, Copenhagen, Denmark
- The National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Henrik Enghusen-Poulsen
- Laboratory of Clinical Pharmacology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center, Gentofte, Denmark
| | - Lars Køber
- Department of Cardiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Christian Torp-Pedersen
- Unit of Epidemiology and biostatistics, Aalborg University Hospital, Aalborg, Denmark
- Department of Health Science and Technology, Aalborg University Hospital, Aalborg, Denmark
| | - Peter E. Weeke
- Department of Cardiology, Bispebjerg and Frederiksberg Hospital, Bispebjerg, Denmark
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
19
|
Pezzolo E, Naldi L. The relationship between smoking, psoriasis and psoriatic arthritis. Expert Rev Clin Immunol 2018; 15:41-48. [DOI: 10.1080/1744666x.2019.1543591] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Elena Pezzolo
- Study Centre of the Italian Group for Epidemiologic Research in Dermatology (GISED), Bergamo, Italy
| | - Luigi Naldi
- Study Centre of the Italian Group for Epidemiologic Research in Dermatology (GISED), Bergamo, Italy
- Department of Dermatology, Ospedale san Bortolo, Vicenza, Italy
| |
Collapse
|
20
|
Pérez‐Morales R, González‐Zamora A, González‐Delgado MF, Calleros Rincón EY, Olivas Calderón EH, Martínez‐Ramírez OC, Rubio J. CHRNA3
rs1051730 and
CHRNA5
rs16969968 polymorphisms are associated with heavy smoking, lung cancer, and chronic obstructive pulmonary disease in a mexican population. Ann Hum Genet 2018; 82:415-424. [DOI: 10.1111/ahg.12264] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Rebeca Pérez‐Morales
- Laboratorio de Biología Celulary Molecular Facultad de Ciencias Químicas, Universidad Juárez del Estado de Durango Durango México
| | - Alberto González‐Zamora
- Laboratorio de Biología Evolutiva. Facultad de Ciencias Biológicas Universidad Juárez del Estado de Durango Durango México
| | - María Fernanda González‐Delgado
- Laboratorio de Biología Celulary Molecular Facultad de Ciencias Químicas, Universidad Juárez del Estado de Durango Durango México
| | - Esperanza Y. Calleros Rincón
- Laboratorio de Biología Celulary Molecular Facultad de Ciencias Químicas, Universidad Juárez del Estado de Durango Durango México
| | - Edgar H. Olivas Calderón
- Laboratorio de Biología Celulary Molecular Facultad de Ciencias Químicas, Universidad Juárez del Estado de Durango Durango México
| | | | - Julieta Rubio
- Departamento de Medicina Genómica y Toxicología Ambiental Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México Ciudad de México México
| |
Collapse
|
21
|
Salloum NC, Buchalter ELF, Chanani S, Espejo G, Ismail MS, Laine RO, Nageeb M, Srivastava AB, Trapp N, Trillo L, Vance E, Wenzinger M, Hartz SM, David SP, Chen LS. From genes to treatments: a systematic review of the pharmacogenetics in smoking cessation. Pharmacogenomics 2018; 19:861-871. [PMID: 29914292 PMCID: PMC6219447 DOI: 10.2217/pgs-2018-0023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 04/30/2018] [Indexed: 12/12/2022] Open
Abstract
Smoking cessation treatment outcomes may be heavily influenced by genetic variations among smokers. Therefore, identifying specific variants that affect response to different pharmacotherapies is of major interest to the field. In the current study, we systematically review all studies published in or after the year 1990 which examined one or more gene-drug interactions for smoking cessation treatment. Out of 644 citations, 46 articles met the inclusion criteria for the systematic review. We summarize evidence on several genetic polymorphisms (CHRNA5-A3-B4, CYP2A6, DBH, CHRNA4, COMT, DRD2, DRD4 and CYP2B6) and their potential moderating pharamacotherarpy effects on patient cessation efficacy rates. These findings are promising and call for further research to demonstrate the effectiveness of genetic testing in personalizing treatment decision-making and improving outcome.
Collapse
Affiliation(s)
- Naji C Salloum
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Erica LF Buchalter
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Swati Chanani
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Gemma Espejo
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Mahjabeen S Ismail
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Randy O Laine
- Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Maysaa Nageeb
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - A Benjamin Srivastava
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Nicholas Trapp
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Ludwig Trillo
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Erica Vance
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Michael Wenzinger
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Sarah M Hartz
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Sean P David
- Department of Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Li-Shiun Chen
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
22
|
Obeidat M, Zhou G, Li X, Hansel NN, Rafaels N, Mathias R, Ruczinski I, Beaty TH, Barnes KC, Paré PD, Sin DD. The genetics of smoking in individuals with chronic obstructive pulmonary disease. Respir Res 2018; 19:59. [PMID: 29631575 PMCID: PMC5892035 DOI: 10.1186/s12931-018-0762-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/27/2018] [Indexed: 11/10/2022] Open
Abstract
Background Smoking is the principal modifiable environmental risk factor for chronic obstructive pulmonary disease (COPD) which affects 300 million people and is the 3rd leading cause of death worldwide. Most of the genetic studies of smoking have relied on self-reported smoking status which is vulnerable to reporting and recall bias. Using data from the Lung Health Study (LHS), we sought to identify genetic variants associated with quantitative smoking and cessation in individuals with mild to moderate COPD. Methods The LHS is a longitudinal multicenter study of mild-to-moderate COPD subjects who were all smokers at recruitment. We performed genome-wide association studies (GWASs) for salivary cotinine (n = 4024), exhaled carbon monoxide (eCO) (n = 2854), cigarettes per day (CPD) (n = 2706) and smoking cessation at year 5 follow-up (n = 717 quitters and 2175 smokers). The GWAS analyses were adjusted for age, gender, and genetic principal components. Results For cotinine levels, SNPs near UGT2B10 gene achieved genome-wide significance (i.e. P < 5 × 10− 8) with top SNP rs10023464, P = 1.27 × 10− 11. For eCO levels, one significant SNP was identified which mapped to the CHRNA3 gene (rs12914385, P = 2.38 × 10− 8). A borderline region mapping to KCNMA1 gene was associated with smoking cessation (rs207675, P = 5.95 × 10− 8). Of the identified loci, only the CHRNA3/5 locus showed significant associations with lung function but only in heavy smokers. No regions met genome-wide significance for CPD. Conclusion The study demonstrates that using objective measures of smoking such as eCO and/or salivary cotinine can more precisely capture the genetic contribution to multiple aspects of smoking behaviour. The KCNMA1 gene association with smoking cessation may represent a potential therapeutic target and warrants further studies. Trial registration The Lung Health Study ClinicalTrials.gov Identifier: NCT00000568. Date of registration: October 28, 1999. Electronic supplementary material The online version of this article (10.1186/s12931-018-0762-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ma'en Obeidat
- The University of British Columbia Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.
| | - Guohai Zhou
- The University of British Columbia Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada
| | - Xuan Li
- The University of British Columbia Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada
| | - Nadia N Hansel
- Pulmonary and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas Rafaels
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Rasika Mathias
- Division of Genetic Epidemiology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Ingo Ruczinski
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Terri H Beaty
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Kathleen C Barnes
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Peter D Paré
- The University of British Columbia Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Respiratory Division, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Don D Sin
- The University of British Columbia Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Respiratory Division, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
23
|
Saccone NL, Baurley JW, Bergen AW, David SP, Elliott HR, Foreman MG, Kaprio J, Piasecki TM, Relton CL, Zawertailo L, Bierut LJ, Tyndale RF, Chen LS, Genetics and Treatment Networks of the Society for Research on Nicotine and Tobacco (SRNT). The Value of Biosamples in Smoking Cessation Trials: A Review of Genetic, Metabolomic, and Epigenetic Findings. Nicotine Tob Res 2018; 20:403-413. [PMID: 28472521 PMCID: PMC5896536 DOI: 10.1093/ntr/ntx096] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 05/01/2017] [Indexed: 02/03/2023]
Abstract
Introduction Human genetic research has succeeded in definitively identifying multiple genetic variants associated with risk for nicotine dependence and heavy smoking. To build on these advances, and to aid in reducing the prevalence of smoking and its consequent health harms, the next frontier is to identify genetic predictors of successful smoking cessation and also of the efficacy of smoking cessation treatments ("pharmacogenomics"). More broadly, additional biomarkers that can be quantified from biosamples also promise to aid "Precision Medicine" and the personalization of treatment, both pharmacological and behavioral. Aims and Methods To motivate ongoing and future efforts, here we review several compelling genetic and biomarker findings related to smoking cessation and treatment. Results These Key results involve genetic variants in the nicotinic receptor subunit gene CHRNA5, variants in the nicotine metabolism gene CYP2A6, and the nicotine metabolite ratio. We also summarize reports of epigenetic changes related to smoking behavior. Conclusions The results to date demonstrate the value and utility of data generated from biosamples in clinical treatment trial settings. This article cross-references a companion paper in this issue that provides practical guidance on how to incorporate biosample collection into a planned clinical trial and discusses avenues for harmonizing data and fostering consortium-based, collaborative research on the pharmacogenomics of smoking cessation. Implications Evidence is emerging that certain genotypes and biomarkers are associated with smoking cessation success and efficacy of smoking cessation treatments. We review key findings that open potential avenues for personalizing smoking cessation treatment according to an individual's genetic or metabolic profile. These results provide important incentive for smoking cessation researchers to collect biosamples and perform genotyping in research studies and clinical trials.
Collapse
Affiliation(s)
- Nancy L Saccone
- Department of Genetics and Division of Biostatistics, Washington University School of Medicine, St. Louis, MO
| | | | | | - Sean P David
- Department of Medicine, Stanford University, Stanford, CA
| | - Hannah R Elliott
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Marilyn G Foreman
- Pulmonary and Critical Care Medicine, Morehouse School of Medicine, Atlanta, GA
| | - Jaakko Kaprio
- Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - Thomas M Piasecki
- Department of Psychological Sciences, University of Missouri, Columbia, MO
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Laurie Zawertailo
- Nicotine Dependence Service, Centre for Addiction and Mental Health, and Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Laura J Bierut
- Siteman Cancer Center, Institute of Public Health, and Department of Psychiatry, Washington University School of Medicine, St. Louis, MO
| | - Rachel F Tyndale
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, and Departments of Pharmacology & Toxicology and Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Li-Shiun Chen
- Siteman Cancer Center, Institute of Public Health, and Department of Psychiatry, Washington University School of Medicine, St. Louis, MO
| | | |
Collapse
|
24
|
Lang BM, Biedermann L, van Haaften WT, de Vallière C, Schuurmans M, Begré S, Zeitz J, Scharl M, Turina M, Greuter T, Schreiner P, Heinrich H, Kuntzen T, Vavricka SR, Rogler G, Beerenwinkel N, Misselwitz B. Genetic polymorphisms associated with smoking behaviour predict the risk of surgery in patients with Crohn's disease. Aliment Pharmacol Ther 2018; 47:55-66. [PMID: 29052254 DOI: 10.1111/apt.14378] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/04/2017] [Accepted: 09/22/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Smoking is a strong environmental factor leading to adverse outcomes in Crohn's disease, but a more benign course in ulcerative colitis. Several single nucleotide polymorphisms (SNPs) are associated with smoking quantity and behaviour. AIM To assess whether smoking-associated SNPs interact with smoking to influence the clinical course of inflammatory bowel diseases. METHODS Genetic and prospectively obtained clinical data from 1434 Swiss inflammatory bowel disease cohort patients (821 Crohn's disease and 613 ulcerative colitis) were analysed. Six SNPs associated with smoking quantity and behaviour (rs588765, rs1051730, rs1329650, rs4105144, rs6474412 and rs3733829) were combined to form a risk score (range: 0-12) by adding the number of risk alleles. We calculated multivariate models for smoking, risk of surgery, fistula, Crohn's disease location and ulcerative colitis disease extent. RESULTS In Crohn's disease patients who smoke, the number of surgeries was associated with the genetic risk score. This translates to a predicted 3.5-fold (95% confidence interval: 2.4- to 5.7-fold, P<.0001) higher number of surgical procedures in smokers with 12 risk alleles than individuals with the lowest risk. Patients with a risk score >7 had a significantly shorter time to first intestinal surgery. The genetic risk score did not predict surgery in ulcerative colitis or occurrence of fistulae in Crohn's disease. SNP rs6265 was associated with ileal disease in Crohn's disease (P<.05) and proctitis in ulcerative colitis (P<.05). CONCLUSIONS SNPs associated with smoking quantity is associated with an increased risk for surgery in Crohn's disease patients who smoke. Our data provide an example of genetics interacting with the environment to influence the disease course of inflammatory bowel disease.
Collapse
Affiliation(s)
- B M Lang
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - L Biedermann
- Division of Gastroenterology, University Hospital Zurich (USZ) and Zurich University, Zurich, Switzerland
| | - W T van Haaften
- Division of Gastroenterology, University Hospital Zurich (USZ) and Zurich University, Zurich, Switzerland.,Department of Gastroenterology and Hepatology, University Medical Center Groningen, Groningen, The Netherlands
| | - C de Vallière
- Division of Gastroenterology, University Hospital Zurich (USZ) and Zurich University, Zurich, Switzerland
| | - M Schuurmans
- Division of Pneumology, University Hospital Zurich (USZ) and Zurich University, Zurich, Switzerland
| | - S Begré
- Hohenegg Hospital, Meilen, Switzerland
| | - J Zeitz
- Division of Gastroenterology, University Hospital Zurich (USZ) and Zurich University, Zurich, Switzerland
| | - M Scharl
- Division of Gastroenterology, University Hospital Zurich (USZ) and Zurich University, Zurich, Switzerland
| | - M Turina
- Division of Visceral Surgery, University Hospital Zurich (USZ), Zurich, Switzerland
| | - T Greuter
- Division of Gastroenterology, University Hospital Zurich (USZ) and Zurich University, Zurich, Switzerland
| | - P Schreiner
- Division of Gastroenterology, University Hospital Zurich (USZ) and Zurich University, Zurich, Switzerland
| | - H Heinrich
- Division of Gastroenterology, University Hospital Zurich (USZ) and Zurich University, Zurich, Switzerland
| | - T Kuntzen
- Division of Gastroenterology, University Hospital Zurich (USZ) and Zurich University, Zurich, Switzerland
| | - S R Vavricka
- Division of Gastroenterology, Triemli Hospital Zurich, Zürich, Switzerland
| | - G Rogler
- Division of Gastroenterology, University Hospital Zurich (USZ) and Zurich University, Zurich, Switzerland
| | - N Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - B Misselwitz
- Division of Gastroenterology, University Hospital Zurich (USZ) and Zurich University, Zurich, Switzerland
| | | |
Collapse
|
25
|
Maes HH, Prom-Wormley E, Eaves LJ, Rhee SH, Hewitt JK, Young S, Corley R, McGue M, Iacono WG, Legrand L, Samek DR, Murrelle EL, Silberg JL, Miles DR, Schieken RM, Beunen GP, Thomis M, Rose RJ, Dick DM, Boomsma DI, Bartels M, Vink JM, Lichtenstein P, White V, Kaprio J, Neale MC. A Genetic Epidemiological Mega Analysis of Smoking Initiation in Adolescents. Nicotine Tob Res 2017; 19:401-409. [PMID: 27807125 DOI: 10.1093/ntr/ntw294] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 10/28/2016] [Indexed: 12/12/2022]
Abstract
Introduction Previous studies in adolescents were not adequately powered to accurately disentangle genetic and environmental influences on smoking initiation (SI) across adolescence. Methods Mega-analysis of pooled genetically informative data on SI was performed, with structural equation modeling, to test equality of prevalence and correlations across cultural backgrounds, and to estimate the significance and effect size of genetic and environmental effects according to the classical twin study, in adolescent male and female twins from same-sex and opposite-sex twin pairs (N = 19 313 pairs) between ages 10 and 19, with 76 358 longitudinal assessments between 1983 and 2007, from 11 population-based twin samples from the United States, Europe, and Australia. Results Although prevalences differed between samples, twin correlations did not, suggesting similar etiology of SI across developed countries. The estimate of additive genetic contributions to liability of SI increased from approximately 15% to 45% from ages 13 to 19. Correspondingly, shared environmental factors accounted for a substantial proportion of variance in liability to SI at age 13 (70%) and gradually less by age 19 (40%). Conclusions Both additive genetic and shared environmental factors significantly contribute to variance in SI throughout adolescence. The present study, the largest genetic epidemiological study on SI to date, found consistent results across 11 studies for the etiology of SI. Environmental factors, especially those shared by siblings in a family, primarily influence SI variance in early adolescence, while an increasing role of genetic factors is seen at later ages, which has important implications for prevention strategies. Implications This is the first study to find evidence of genetic factors in liability to SI at ages as young as 12. It also shows the strongest evidence to date for decay of effects of the shared environment from early adolescence to young adulthood. We found remarkable consistency of twin correlations across studies reflecting similar etiology of liability to initiate smoking across different cultures and time periods. Thus familial factors strongly contribute to individual differences in who starts to smoke with a gradual increase in the impact of genetic factors and a corresponding decrease in that of the shared environment.
Collapse
Affiliation(s)
- Hermine H Maes
- Department of Human and Molecular Genetics, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA.,Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA.,Department of Kinesiology, KU Leuven, Leuven, Belgium
| | - Elizabeth Prom-Wormley
- Department of Human and Molecular Genetics, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA.,Division of Epidemiology, Department of Family Medicine and Population Health, Virginia Commonwealth University, Richmond, VA
| | - Lindon J Eaves
- Department of Human and Molecular Genetics, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA.,Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA
| | - Soo Hyun Rhee
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO
| | - John K Hewitt
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO
| | - Susan Young
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO.,Department of Psychiatry, University of Colorado, Denver, CO
| | - Robin Corley
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO
| | - Matt McGue
- Department of Psychology, University of Minnesota, Minneapolis, MN
| | - William G Iacono
- Department of Psychology, University of Minnesota, Minneapolis, MN
| | - Lisa Legrand
- Department of Psychology, University of Minnesota, Minneapolis, MN
| | - Diana R Samek
- Department of Psychology, University of Minnesota, Minneapolis, MN.,Human Development and Family Studies Department, Auburn University, Auburn, AL
| | | | - Judy L Silberg
- Department of Human and Molecular Genetics, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA.,Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA
| | - Donna R Miles
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Richard M Schieken
- Department of Pediatric Cardiology, Virginia Commonwealth University, Richmond, VA
| | | | | | - Richard J Rose
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN
| | - Danielle M Dick
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA.,Department of Psychology, Virginia Commonwealth University, Richmond, VA
| | - Dorret I Boomsma
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Meike Bartels
- Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| | - Jacqueline M Vink
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands.,Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| | - Paul Lichtenstein
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Victoria White
- Centre for Behavioural Research in Cancer, Cancer Council Victoria, Melbourne, Australia
| | - Jaakko Kaprio
- Department of Public Health and Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland.,Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Michael C Neale
- Department of Human and Molecular Genetics, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA.,Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
26
|
A little rein on addiction. Semin Cell Dev Biol 2017; 78:120-129. [PMID: 28986065 DOI: 10.1016/j.semcdb.2017.09.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 09/18/2017] [Accepted: 09/20/2017] [Indexed: 10/18/2022]
Abstract
Rewarding and aversive experiences influence emotions, motivate specific behaviors, and modify future action in animals. Multiple conserved vertebrate neural circuits have been discovered that act in a species-specific manner to reinforce behaviors that are rewarding, while attenuating those with an adverse outcome. A growing body of research now suggests that malfunction of the same circuits is an underlying cause for many human disorders and mental ailments. The habenula (Latin for "little rein") complex, an epithalamic structure that regulates midbrain monoaminergic activity has emerged in recent years as one such region in the vertebrate brain that modulates behavior. Its dysfunction, on the other hand, is implicated in a spectrum of psychiatric disorders in humans such as schizophrenia, depression and addiction. Here, I review the progress in identification of potential mechanisms involving the habenula in addiction.
Collapse
|
27
|
Gupta R, Qaiser B, He L, Hiekkalinna TS, Zheutlin AB, Therman S, Ollikainen M, Ripatti S, Perola M, Salomaa V, Milani L, Cannon TD, Madden PAF, Korhonen T, Kaprio J, Loukola A. Neuregulin signaling pathway in smoking behavior. Transl Psychiatry 2017; 7:e1212. [PMID: 28892072 PMCID: PMC5611747 DOI: 10.1038/tp.2017.183] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 06/26/2017] [Accepted: 07/06/2017] [Indexed: 12/23/2022] Open
Abstract
Understanding molecular processes that link comorbid traits such as addictions and mental disorders can provide novel therapeutic targets. Neuregulin signaling pathway (NSP) has previously been implicated in schizophrenia, a neurodevelopmental disorder with high comorbidity to smoking. Using a Finnish twin family sample, we have previously detected association between nicotine dependence and ERBB4 (a neuregulin receptor), and linkage for smoking initiation at the ERBB4 locus on 2q33. Further, Neuregulin3 has recently been shown to associate with nicotine withdrawal in a behavioral mouse model. In this study, we scrutinized association and linkage between 15 036 common, low frequency and rare genetic variants in 10 NSP genes and phenotypes encompassing smoking and alcohol use. Using the Finnish twin family sample (N=1998 from 740 families), we detected 66 variants (representing 23 LD blocks) significantly associated (false discovery rate P<0.05) with smoking initiation, nicotine dependence and nicotine withdrawal. We comprehensively annotated the associated variants using expression (eQTL) and methylation quantitative trait loci (meQTL) analyses in a Finnish population sample. Among the 66 variants, we identified 25 eQTLs (in NRG1 and ERBB4), 22 meQTLs (in NRG3, ERBB4 and PSENEN), a missense variant in NRG1 (rs113317778) and a splicing disruption variant in ERBB4 (rs13385826). Majority of the QTLs in blood were replicated in silico using publicly available databases, with additional QTLs observed in brain. In conclusion, our results support the involvement of NSP in smoking behavior but not in alcohol use and abuse, and disclose functional potential for 56 of the 66 associated single-nucleotide polymorphism.
Collapse
Affiliation(s)
- R Gupta
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - B Qaiser
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - L He
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, USA
| | - T S Hiekkalinna
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | - A B Zheutlin
- Department of Psychology, Yale University, New Haven, CT, USA
| | - S Therman
- National Institute for Health and Welfare, Helsinki, Finland
| | - M Ollikainen
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - S Ripatti
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - M Perola
- National Institute for Health and Welfare, Helsinki, Finland
| | - V Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - L Milani
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - T D Cannon
- Department of Psychology, Yale University, New Haven, CT, USA
| | - P A F Madden
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - T Korhonen
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
- National Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - J Kaprio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - A Loukola
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| |
Collapse
|
28
|
Begum F, Ruczinski I, Hokanson JE, Lutz SM, Parker MM, Cho MH, Hetmanski JB, Scharpf RB, Crapo JD, Silverman EK, Beaty TH. Hemizygous Deletion on Chromosome 3p26.1 Is Associated with Heavy Smoking among African American Subjects in the COPDGene Study. PLoS One 2016; 11:e0164134. [PMID: 27711239 PMCID: PMC5053531 DOI: 10.1371/journal.pone.0164134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/20/2016] [Indexed: 02/04/2023] Open
Abstract
Many well-powered genome-wide association studies have identified genetic determinants of self-reported smoking behaviors and measures of nicotine dependence, but most have not considered the role of structural variants, such as copy number variation (CNVs), influencing these phenotypes. Here, we included 2,889 African American and 6,187 non-Hispanic White subjects from the COPDGene cohort (http://www.copdgene.org) to carefully investigate the role of polymorphic CNVs across the genome on various measures of smoking behavior. We identified a CNV component (a hemizygous deletion) on chromosome 3p26.1 associated with two quantitative phenotypes related to smoking behavior among African Americans. This polymorphic hemizygous deletion is significantly associated with pack-years and cigarettes smoked per day among African American subjects in the COPDGene study. We sought evidence of replication in African Americans from the population based Atherosclerosis Risk in Communities (ARIC) study. While we observed similar CNV counts, the extent of exposure to cigarette smoking among ARIC subjects was quite different and the smaller sample size of heavy smokers in ARIC severely limited statistical power, so we were unable to replicate our findings from the COPDGene cohort. But meta-analyses of COPDGene and ARIC study subjects strengthened our association signal. However, a few linkage studies have reported suggestive linkage to the 3p26.1 region, and a few genome-wide association studies (GWAS) have reported markers in the gene (GRM7) nearest to this 3p26.1 area of polymorphic deletions are associated with measures of nicotine dependence among subjects of European ancestry.
Collapse
Affiliation(s)
- Ferdouse Begum
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - John E. Hokanson
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, United States of America
| | - Sharon M. Lutz
- Department of Biostatisitics and Informatics, Colorado School of Public Health, Aurora, Colorado, United States of America
| | - Margaret M. Parker
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Michael H. Cho
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jacqueline B. Hetmanski
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Robert B. Scharpf
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - James D. Crapo
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Edwin K. Silverman
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Terri H. Beaty
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
29
|
Abstract
Tobacco use remains the leading cause of preventable death in the US, emphasizing the need to understand which genes and environments are involved in the establishment of cigarette use behaviors. However, to date, no comprehensive review of the influence of genes, the environment, and their interaction on cigarette use exists. This narrative review provides a description of gene variants and environmental factors associated with cigarette use, as well as an overview of studies investigating gene-environment interaction (GxE) in cigarette use. GxE studies of cigarette use have been useful in demonstrating that the influence of genes changes as a function of both the phenotype being measured and the environment. However, it is difficult to determine how the effect of genes contributing to different phenotypes of cigarette use changes as a function of the environment. This suggests the need for more studies of GxE, to parse out the effects of genes and the environment across the development of cigarette use phenotypes, which may help to inform potential prevention and intervention efforts aimed at reducing the prevalence of cigarette use. Key Messages No comprehensive reviews of the influence of genes, the environment, and their interaction on cigarette use exist currently. The influence of genes may change as a function of the environment and the phenotype being measured. It is difficult to determine how the effect of genes contributing to different phenotypes of cigarette use changes according to environmental context, suggesting the need for more studies of gene-environment interaction related to cigarette use to parse out effects.
Collapse
Affiliation(s)
- Elizabeth Do
- a Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University , Richmond , VA , USA ;,b Center for Clinical and Translational Research , Virginia Commonwealth University , Richmond , VA , USA
| | - Hermine Maes
- a Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University , Richmond , VA , USA ;,c Department of Human and Molecular Genetics , Virginia Commonwealth University , Richmond , VA , USA ;,d Department of Psychiatry , Virginia Commonwealth University , Richmond , VA , USA ;,e Massey Cancer Center , Virginia Commonwealth University , Richmond , VA , USA
| |
Collapse
|
30
|
Abstract
Smoking is a complex environmental exposure influenced by genetic, environmental, and social factors. Nicotine is the principal alkaloid in tobacco that mediates the addicting effects of tobacco products. Tobacco is a mixture of more than 7,000 chemicals, and smoking is recognized as a risk factor for many diseases in humans, including cardiovascular and pulmonary disease and several cancers, and is the single most preventable cause of mortality worldwide. A number of inflammatory immune-related conditions have been associated with smoking, including psoriasis. Smoking affects the onset of psoriasis. In a pooled analysis of 25 case-control studies, the odds ratio of psoriasis among smokers was 1.78 (95% confidence interval [CI]: 1.53-2.06). A dose-effect relationship is also documented. In a pooled analysis of three cohort studies, the risk of incident psoriasis was 1.81 (95% CI: 1.38-2.36) in those who smoked 1-14 cigarettes per day, and 2.29 (95% CI: 1.74-3.01) in those who smoked ≥25 cigarettes per day. Smoking also impacts on the clinical severity of psoriasis, its response to treatment, and explains some of the associated comorbidities, eg, cardiovascular disease, inflammatory bowel disease, and several cancers (especially those of the respiratory tract). Data on the role of smoking in psoriatic arthritis are less consistent compared with those concerning psoriasis. Several pathophysiological mechanisms may explain the association of psoriasis with smoking, including oxidative stress, interaction with signaling pathways active in psoriasis, and vascular influences. In conclusion, psoriasis is just one of the many diseases associated with smoking, but it is visible and disabling. Dermatologists could play a major role in reducing the health burden of smoking by influencing the patients to change their behavior.
Collapse
Affiliation(s)
- Luigi Naldi
- Department of Dermatology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
- Study Centre of the Italian Group for Epidemiologic Research in Dermatology (GISED), Bergamo, Italy
| |
Collapse
|
31
|
Clark SL, McClay JL, Adkins DE, Aberg KA, Kumar G, Nerella S, Xie L, Collins AL, Crowley JJ, Quakenbush CR, Hillard CE, Gao G, Shabalin AA, Peterson RE, Copeland WE, Silberg JL, Maes H, Sullivan PF, Costello EJ, van den Oord EJ. Deep Sequencing of Three Loci Implicated in Large-Scale Genome-Wide Association Study Smoking Meta-Analyses. Nicotine Tob Res 2016; 18:626-31. [PMID: 26283763 PMCID: PMC5942610 DOI: 10.1093/ntr/ntv166] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 07/17/2015] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Genome-wide association study meta-analyses have robustly implicated three loci that affect susceptibility for smoking: CHRNA5\CHRNA3\CHRNB4, CHRNB3\CHRNA6 and EGLN2\CYP2A6. Functional follow-up studies of these loci are needed to provide insight into biological mechanisms. However, these efforts have been hampered by a lack of knowledge about the specific causal variant(s) involved. In this study, we prioritized variants in terms of the likelihood they account for the reported associations. METHODS We employed targeted capture of the CHRNA5\CHRNA3\CHRNB4, CHRNB3\CHRNA6, and EGLN2\CYP2A6 loci and flanking regions followed by next-generation deep sequencing (mean coverage 78×) to capture genomic variation in 363 individuals. We performed single locus tests to determine if any single variant accounts for the association, and examined if sets of (rare) variants that overlapped with biologically meaningful annotations account for the associations. RESULTS In total, we investigated 963 variants, of which 71.1% were rare (minor allele frequency < 0.01), 6.02% were insertion/deletions, and 51.7% were catalogued in dbSNP141. The single variant results showed that no variant fully accounts for the association in any region. In the variant set results, CHRNB4 accounts for most of the signal with significant sets consisting of directly damaging variants. CHRNA6 explains most of the signal in the CHRNB3\CHRNA6 locus with significant sets indicating a regulatory role for CHRNA6. Significant sets in CYP2A6 involved directly damaging variants while the significant variant sets suggested a regulatory role for EGLN2. CONCLUSIONS We found that multiple variants implicating multiple processes explain the signal. Some variants can be prioritized for functional follow-up.
Collapse
Affiliation(s)
- Shaunna L Clark
- Center for Biomarker Research and Precision Medicine, School of Pharmacy, Virginia Commonwealth University, Richmond, VA;
| | - Joseph L McClay
- Center for Biomarker Research and Precision Medicine, School of Pharmacy, Virginia Commonwealth University, Richmond, VA
| | - Daniel E Adkins
- Center for Biomarker Research and Precision Medicine, School of Pharmacy, Virginia Commonwealth University, Richmond, VA
| | - Karolina A Aberg
- Center for Biomarker Research and Precision Medicine, School of Pharmacy, Virginia Commonwealth University, Richmond, VA
| | - Gaurav Kumar
- Center for Biomarker Research and Precision Medicine, School of Pharmacy, Virginia Commonwealth University, Richmond, VA
| | - Sri Nerella
- Center for Biomarker Research and Precision Medicine, School of Pharmacy, Virginia Commonwealth University, Richmond, VA
| | - Linying Xie
- Center for Biomarker Research and Precision Medicine, School of Pharmacy, Virginia Commonwealth University, Richmond, VA
| | - Ann L Collins
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - James J Crowley
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Corey R Quakenbush
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Christopher E Hillard
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Guimin Gao
- Department of Biostatistics, School of Medicine, Virginia Commonwealth University, Richmond, VA
| | - Andrey A Shabalin
- Center for Biomarker Research and Precision Medicine, School of Pharmacy, Virginia Commonwealth University, Richmond, VA
| | - Roseann E Peterson
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA
| | - William E Copeland
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC
| | - Judy L Silberg
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA
| | - Hermine Maes
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA
| | - Patrick F Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Elizabeth J Costello
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC
| | - Edwin J van den Oord
- Center for Biomarker Research and Precision Medicine, School of Pharmacy, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
32
|
Chromosome 15q25 (CHRNA3-CHRNB4) Variation Indirectly Impacts Lung Cancer Risk in Chinese Males. PLoS One 2016; 11:e0149946. [PMID: 26942719 PMCID: PMC4778880 DOI: 10.1371/journal.pone.0149946] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 02/07/2016] [Indexed: 11/19/2022] Open
Abstract
Introduction Recently, genome-wide association studies (GWAS) in Caucasian populations have identified an association between single nucleotide polymorphisms (SNPs) in the CHRNA5-A3-B4 nicotinic acetylcholine receptor subunit gene cluster on chromosome 15q25, lung cancer risk and smoking behaviors. However, these SNPs are rare in Asians, and there is currently no consensus on whether SNPs in CHRNA5-A3-B4 have a direct or indirect carcinogenic effect through smoking behaviors on lung cancer risk. Though some studies confirmed rs6495308 polymorphisms to be associated with smoking behaviors and lung cancer, no research was conducted in China. Using a case-control study, we decided to investigate the associations between CHRNA3 rs6495308, CHRNB4 rs11072768, smoking behaviors and lung cancer risk, as well as explore whether the two SNPs have a direct or indirect carcinogenic effect on lung cancer. Methods A total of 1025 males were interviewed using a structured questionnaire (204 male lung cancer patients and 821 healthy men) to acquire socio-demographic status and smoking behaviors. Venous blood samples were collected to measure rs6495308 and rs11072768 gene polymorphisms. All subjects were divided into 3 groups: non-smokers, light smokers (1–15 cigarettes per day) and heavy smokers (>15 cigarettes per day). Results Compared to wild genotype, rs6495308 and rs11072768 variant genotypes reported smoking more cigarettes per day and a higher pack-years of smoking (P<0.05). More importantly, among smokers, both rs6495308 CT/TT and rs11072768 GT/GG had a higher risk of lung cancer compared to wild genotype without adjusting for potential confounding factors (OR = 1.36, 95%CI = 1.09–1.95; OR = 1.11, 95%CI = 1.07–1.58 respectively). Furthermore, heavy smokers with rs6495308 or rs11072768 variant genotypes have a positive interactive effect on lung cancer after adjustment for potential confounding factors (OR = 1.13, 95%CI = 1.01–3.09; OR = 1.09, 95%CI = 1.01–3.41 respectively). However, No significant associations were found between lung cancer risk and both rs6495308 and rs11072768 genotypes among non-smokers and smokers after adjusting for age, occupation, and education. Conclusion This study confirmed both rs6495308 and rs11072768 gene polymorphisms association with smoking behaviors and had an indirect link between gene polymorphisms and lung cancer risk.
Collapse
|
33
|
Transmission of Smoking across Three Generations in Finland. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 13:74. [PMID: 26712771 PMCID: PMC4730465 DOI: 10.3390/ijerph13010074] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/17/2015] [Accepted: 12/21/2015] [Indexed: 11/30/2022]
Abstract
The influence of parents’ smoking on children’s smoking is well known, but few studies have examined the association between grandparents’ and grandchildren’s smoking. We studied the association between paternal and maternal grandparents’ smoking and their grandchildren’s tobacco use and assessed whether parents’ smoking is a mediator in this process. Data were obtained from a national survey of 12–18-year-old Finns in 2013 (N = 3535, response rate 38%). Logistic regression and mediation analyses were used. Both boys and girls had higher odds for smoking experimentation, daily smoking and other tobacco or tobacco-like product use if their mother, father or any of the four grandparents were current or former smokers. When parents’ and grandparents’ smoking status were included in the same model, grandparents’ smoking generally lost statistical significance. In the mediation analysis, 73% of the total effect of grandparents’ smoking on grandchildren’s daily smoking was mediated through parents’ smoking, 64% on smoking experimentation and 63% on other tobacco or tobacco-like product use. The indirect effect of a mother’s smoking was higher than that of a father’s. To conclude, paternal and maternal grandparents’ smoking increases grandchildren’s tobacco use. The influence is mainly, but not completely, mediated through parents’ smoking.
Collapse
|
34
|
Li H, Li S, Wang Q, Pan L, Jiang F, Yang X, Zhang N, Han M, Jia C. Association of 5-HTTLPR polymorphism with smoking behaviors: A meta-analysis. Physiol Behav 2015; 152:32-40. [DOI: 10.1016/j.physbeh.2015.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 08/11/2015] [Accepted: 09/08/2015] [Indexed: 10/23/2022]
|
35
|
Phillips TJ, Shabani S. An animal model of differential genetic risk for methamphetamine intake. Front Neurosci 2015; 9:327. [PMID: 26441502 PMCID: PMC4585292 DOI: 10.3389/fnins.2015.00327] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/31/2015] [Indexed: 11/13/2022] Open
Abstract
The question of whether genetic factors contribute to risk for methamphetamine (MA) use and dependence has not been intensively investigated. Compared to human populations, genetic animal models offer the advantages of control over genetic family history and drug exposure. Using selective breeding, we created lines of mice that differ in genetic risk for voluntary MA intake and identified the chromosomal addresses of contributory genes. A quantitative trait locus was identified on chromosome 10 that accounts for more than 50% of the genetic variance in MA intake in the selected mouse lines. In addition, behavioral and physiological screening identified differences corresponding with risk for MA intake that have generated hypotheses that are testable in humans. Heightened sensitivity to aversive and certain physiological effects of MA, such as MA-induced reduction in body temperature, are hallmarks of mice bred for low MA intake. Furthermore, unlike MA-avoiding mice, MA-preferring mice are sensitive to rewarding and reinforcing MA effects, and to MA-induced increases in brain extracellular dopamine levels. Gene expression analyses implicate the importance of a network enriched in transcription factor genes, some of which regulate the mu opioid receptor gene, Oprm1, in risk for MA use. Neuroimmune factors appear to play a role in differential response to MA between the mice bred for high and low intake. In addition, chromosome 10 candidate gene studies provide strong support for a trace amine-associated receptor 1 gene, Taar1, polymorphism in risk for MA intake. MA is a trace amine-associated receptor 1 (TAAR1) agonist, and a non-functional Taar1 allele segregates with high MA consumption. Thus, reduced TAAR1 function has the potential to increase risk for MA use. Overall, existing findings support the MA drinking lines as a powerful model for identifying genetic factors involved in determining risk for harmful MA use. Future directions include the development of a binge model of MA intake, examining the effect of withdrawal from chronic MA on MA intake, and studying potential Taar1 gene × gene and gene × environment interactions. These and other studies are intended to improve our genetic model with regard to its translational value to human addiction.
Collapse
Affiliation(s)
- Tamara J. Phillips
- VA Portland Health Care SystemPortland, OR, USA
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science UniversityPortland, OR, USA
| | | |
Collapse
|
36
|
Abstract
It is now well-established that smoking-related behaviours are under a substantial degree of genetic influence. Efforts are now focused on identifying the specific genetic variants which underlie these behaviours. Within this chapter, we introduce a variety of established and emerging methods employed to identify such variants, ranging from candidate gene to whole genome sequencing approaches, and highlight what these techniques have taught us about the genetic architecture of smoking-related behaviours. Further, we discuss how phenotype refinement has developed our understanding of these relationships, affording us insight into the specific mechanisms linking genetic variants to smoking-related behaviours.
Collapse
|
37
|
Genetic influences on smoking and clinical disease. Understanding behavioral and biological pathways with mediation analysis. Ann Am Thorac Soc 2015; 11:1082-3. [PMID: 25237988 DOI: 10.1513/annalsats.201407-315ed] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
38
|
Healthy lifestyles and personal responsibility. J Am Coll Cardiol 2014; 64:1786-8. [PMID: 25443699 DOI: 10.1016/j.jacc.2014.08.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 07/26/2014] [Accepted: 08/03/2014] [Indexed: 11/21/2022]
|
39
|
Taylor AE, Fluharty ME, Bjørngaard JH, Gabrielsen ME, Skorpen F, Marioni RE, Campbell A, Engmann J, Mirza SS, Loukola A, Laatikainen T, Partonen T, Kaakinen M, Ducci F, Cavadino A, Husemoen LLN, Ahluwalia TS, Jacobsen RK, Skaaby T, Ebstrup JF, Mortensen EL, Minica CC, Vink JM, Willemsen G, Marques-Vidal P, Dale CE, Amuzu A, Lennon LT, Lahti J, Palotie A, Räikkönen K, Wong A, Paternoster L, Wong APY, Horwood LJ, Murphy M, Johnstone EC, Kennedy MA, Pausova Z, Paus T, Ben-Shlomo Y, Nohr EA, Kuh D, Kivimaki M, Eriksson JG, Morris RW, Casas JP, Preisig M, Boomsma DI, Linneberg A, Power C, Hyppönen E, Veijola J, Jarvelin MR, Korhonen T, Tiemeier H, Kumari M, Porteous DJ, Hayward C, Romundstad PR, Smith GD, Munafò MR. Investigating the possible causal association of smoking with depression and anxiety using Mendelian randomisation meta-analysis: the CARTA consortium. BMJ Open 2014; 4:e006141. [PMID: 25293386 PMCID: PMC4187451 DOI: 10.1136/bmjopen-2014-006141] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES To investigate whether associations of smoking with depression and anxiety are likely to be causal, using a Mendelian randomisation approach. DESIGN Mendelian randomisation meta-analyses using a genetic variant (rs16969968/rs1051730) as a proxy for smoking heaviness, and observational meta-analyses of the associations of smoking status and smoking heaviness with depression, anxiety and psychological distress. PARTICIPANTS Current, former and never smokers of European ancestry aged ≥16 years from 25 studies in the Consortium for Causal Analysis Research in Tobacco and Alcohol (CARTA). PRIMARY OUTCOME MEASURES Binary definitions of depression, anxiety and psychological distress assessed by clinical interview, symptom scales or self-reported recall of clinician diagnosis. RESULTS The analytic sample included up to 58 176 never smokers, 37 428 former smokers and 32 028 current smokers (total N=127 632). In observational analyses, current smokers had 1.85 times greater odds of depression (95% CI 1.65 to 2.07), 1.71 times greater odds of anxiety (95% CI 1.54 to 1.90) and 1.69 times greater odds of psychological distress (95% CI 1.56 to 1.83) than never smokers. Former smokers also had greater odds of depression, anxiety and psychological distress than never smokers. There was evidence for positive associations of smoking heaviness with depression, anxiety and psychological distress (ORs per cigarette per day: 1.03 (95% CI 1.02 to 1.04), 1.03 (95% CI 1.02 to 1.04) and 1.02 (95% CI 1.02 to 1.03) respectively). In Mendelian randomisation analyses, there was no strong evidence that the minor allele of rs16969968/rs1051730 was associated with depression (OR=1.00, 95% CI 0.95 to 1.05), anxiety (OR=1.02, 95% CI 0.97 to 1.07) or psychological distress (OR=1.02, 95% CI 0.98 to 1.06) in current smokers. Results were similar for former smokers. CONCLUSIONS Findings from Mendelian randomisation analyses do not support a causal role of smoking heaviness in the development of depression and anxiety.
Collapse
Affiliation(s)
- Amy E Taylor
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, UK
- UK Centre for Tobacco and Alcohol Studies, School of Experimental Psychology, University of Bristol, Bristol, UK
| | - Meg E Fluharty
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, UK
- UK Centre for Tobacco and Alcohol Studies, School of Experimental Psychology, University of Bristol, Bristol, UK
| | - Johan H Bjørngaard
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Forensic Department, Research Centre Bröset St. Olav's University Hospital Trondheim, Trondheim, Norway
| | - Maiken Elvestad Gabrielsen
- Faculty of Medicine, Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Frank Skorpen
- Faculty of Medicine, Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Riccardo E Marioni
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Archie Campbell
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Jorgen Engmann
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Saira Saeed Mirza
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Anu Loukola
- University of Helsinki, Hjelt institute, Helsinki, Finland
| | - Tiina Laatikainen
- University of Eastern Finland, Institute of Public Health & Clinical Nutrition, Kuopio, Finland
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
- Hospital District of North Karelia, Joensuu, Finland
| | - Timo Partonen
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland
| | - Marika Kaakinen
- Institute of Health Sciences, FI-90014 University of Oulu, Finland
- Biocenter Oulu, FI-90014 University of Oulu, Finland
| | - Francesca Ducci
- South West London and St George's Mental Health Trust, London, UK
| | - Alana Cavadino
- Population, Policy and Practice, UCL Institute of Child Health, University College London, UK
| | | | - Tarunveer Singh Ahluwalia
- Metabolic Genetics Section, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Copenhagen Prospective Studies on Asthma in Childhood, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Danish Pediatric Asthma Center, Gentofte Hospital, The Capital Region, Copenhagen, Denmark
| | - Rikke Kart Jacobsen
- Research Centre for Prevention and Health, the Capital Region of Denmark, Denmark
| | - Tea Skaaby
- Research Centre for Prevention and Health, the Capital Region of Denmark, Denmark
| | | | - Erik Lykke Mortensen
- Institute of Public Health and Center for Healthy Aging, University of Copenhagen, Denmark
| | - Camelia C Minica
- Department of Biological Psychology, Netherlands Twin Register, VU University, Amsterdam, The Netherlands
| | - Jacqueline M Vink
- Department of Biological Psychology, Netherlands Twin Register, VU University, Amsterdam, The Netherlands
| | - Gonneke Willemsen
- Department of Biological Psychology, Netherlands Twin Register, VU University, Amsterdam, The Netherlands
| | - Pedro Marques-Vidal
- Department of Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Caroline E Dale
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Antoinette Amuzu
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Lucy T Lennon
- Department of Primary Care & Population Health, UCL, London, UK
| | - Jari Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Centre, Helsinki, Finland
| | - Aarno Palotie
- Wellcome Trust Sanger Institute, Cambridge, UK
- The Medical and Population Genomics Program, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Finland
| | | | - Andrew Wong
- MRC Unit for Lifelong Health, Ageing at UCL, UK
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Angelita Pui-Yee Wong
- Department of Psychology, University of Toronto, Toronto, Canada
- Rotman Research Institute, Toronto, Canada
| | - L John Horwood
- Department of Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - Michael Murphy
- Childhood Cancer Research Group, University of Oxford, Oxford, UK
| | | | - Martin A Kennedy
- Department of Pathology, University of Otago, Christchurch, New Zealand
| | - Zdenka Pausova
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Canada
- Hospital for Sick Children, Toronto, Canada
| | - Tomáš Paus
- Rotman Research Institute, Toronto, Canada
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, Canada
| | - Yoav Ben-Shlomo
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Ellen A Nohr
- Institute for Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Diana Kuh
- MRC Unit for Lifelong Health, Ageing at UCL, UK
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Johan G Eriksson
- Folkhälsan Research Centre, Helsinki, Finland
- Department of Medical Genetics, University of Helsinki and University Central Hospital, Helsinki, Finland
- National Institute for Health and Welfare, Finland
- Department of General Practice and Primary health Care, University of Helsinki, Finland
- Unit of General Practice, Helsinki University Central Hospital, Helsinki, Finland
- Vasa Central Hospital, Vasa, Finland
| | | | - Juan P Casas
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
- Institute of Cardiovascular Science, University College London, UK
| | - Martin Preisig
- Department of Psychiatry, Lausanne University Hospital, Prilly, Switzerland
| | - Dorret I Boomsma
- Department of Biological Psychology, Netherlands Twin Register, VU University, Amsterdam, The Netherlands
| | - Allan Linneberg
- Research Centre for Prevention and Health, the Capital Region of Denmark, Denmark
- Department of Clinical Experimental Research, Glostrup University Hospital, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Chris Power
- Population, Policy and Practice, UCL Institute of Child Health, University College London, UK
| | - Elina Hyppönen
- Population, Policy and Practice, UCL Institute of Child Health, University College London, UK
- School of Population Health and Sansom Institute, University of South Australia, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Juha Veijola
- Department of Psychiatry, Oulu University Hospital, Oulu, Finland
| | - Marjo-Riitta Jarvelin
- Institute of Health Sciences, FI-90014 University of Oulu, Finland
- Biocenter Oulu, FI-90014 University of Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College London, UK
- Unit of Primary Care, Oulu University Hospital, Oulu, Finland
- Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland
| | - Tellervo Korhonen
- University of Helsinki, Hjelt institute, Helsinki, Finland
- University of Eastern Finland, Institute of Public Health & Clinical Nutrition, Kuopio, Finland
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland
| | - Henning Tiemeier
- Department of Epidemiology and Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Meena Kumari
- Department of Epidemiology and Public Health, University College London, London, UK
| | - David J Porteous
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Pål R Romundstad
- Department of Public Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, UK
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Marcus R Munafò
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, UK
- UK Centre for Tobacco and Alcohol Studies, School of Experimental Psychology, University of Bristol, Bristol, UK
| |
Collapse
|
40
|
Taylor AE, Munafò MR. Commentary: Does mortality from smoking have implications for future Mendelian randomization studies? Int J Epidemiol 2014; 43:1483-6. [PMID: 25125581 PMCID: PMC4190520 DOI: 10.1093/ije/dyu151] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Amy E Taylor
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, UK Centre for Tobacco and Alcohol Research Studies, School of Experimental Psychology, University of Bristol, Bristol, UK
| | - Marcus R Munafò
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, UK Centre for Tobacco and Alcohol Research Studies, School of Experimental Psychology, University of Bristol, Bristol, UK
| |
Collapse
|