51
|
Kaur G, Batra S. Regulation of DNA methylation signatures on NF-κB and STAT3 pathway genes and TET activity in cigarette smoke extract-challenged cells/COPD exacerbation model in vitro. Cell Biol Toxicol 2020; 36:459-480. [PMID: 32342329 DOI: 10.1007/s10565-020-09522-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/19/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a global health problem. Currently, there is a lack of knowledge about the pathobiology of this disease and available therapies are ineffective. Cigarette smoking is the leading cause of COPD; however, not all smokers develop COPD. Exacerbations of COPD caused by microbes are common and detrimental. Approximately 20-50% of patient exacerbations are caused by bacterial colonization in the lower airways. It is generally accepted that epigenetic mechanisms, especially DNA methylation, play an important role during progression of COPD. Thus, we hypothesized that DNA methylation patterns vary significantly following smoke exposure and during exacerbations caused by bacterial infections. To test our hypothesis, we used an in vitro study model that mimics COPD exacerbations and performed extensive studies to understand the role of CpG promoter methylation of NF-κB and STAT3-mediated pathway genes. Both NF-κB and STAT3 transcription factors play critical roles in orchestrating inflammatory responses during cigarette smoke exposure. In brief, human lung adenocarcinoma cells with type II alveolar epithelium characteristics (A549) were challenged with cigarette smoke extract (CSE) or DMSO (control) followed by a 3-h challenge with bacterial lipopolysaccharide (LPS; from Pseudomonas aeruginosa) prior to the termination of CSE exposure (COPD exacerbation group). The production of cytokines/chemokines, regulation of transcription factors, and DNA methylation of specific genes were then assessed. We also studied changes in the expression and activity of ten-eleven translocases (TETs), the enzymes responsible for DNA demethylation, and assessed their role in regulating DNA methylation in the CSE-challenged group. RESULTS There was a significant increase in the release of cytokines/chemokines (IL-8, MCP-1, IL-6 and CCL5) in the COPD exacerbation group as compared to the control group. Hypomethylation of NF-κB-mediated pathway genes correlated with their induction in our COPD exacerbation study model. Further, we observed an important role of TET1/2 in regulating the DNA methylation of NF-κB, STAT3, IKK, and NIK genes and cytokine/chemokine production by A549 cells during CSE challenge. CONCLUSIONS Studies to further define the role of TETs in CSE-mediated epigenetic regulation may lead to the development of better and more effective therapeutic intervention strategies for COPD.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Laboratory of Pulmonary Immunotoxicology, Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Sanjay Batra
- Laboratory of Pulmonary Immunotoxicology, Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA, 70813, USA.
| |
Collapse
|
52
|
Epigenome-Wide Tobacco-Related Methylation Signature Identification and Their Multilevel Regulatory Network Inference for Lung Adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2471915. [PMID: 32420331 PMCID: PMC7201762 DOI: 10.1155/2020/2471915] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/04/2020] [Indexed: 12/12/2022]
Abstract
Tobacco exposure is one of the major risks for the initiation and progress of lung cancer. The exact corresponding mechanisms, however, are mainly unknown. Recently, a growing body of evidence has been collected supporting the involvement of DNA methylation in the regulation of gene expression in cancer cells. The identification of tobacco-related signature methylation probes and the analysis of their regulatory networks at different molecular levels may be of a great help for understanding tobacco-related tumorigenesis. Three independent lung adenocarcinoma (LUAD) datasets were used to train and validate the tobacco exposure pattern classification model. A deep selecting method was proposed and used to identify methylation signature probes from hundreds of thousands of the whole epigenome probes. Then, BIMC (biweight midcorrelation coefficient) algorithm, SRC (Spearman's rank correlation) analysis, and shortest path tracing method were explored to identify associated genes at gene regulation level and protein-protein interaction level, respectively. Afterwards, the KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analysis and GO (Gene Ontology) enrichment analysis were used to analyze their molecular functions and associated pathways. 105 probes were identified as tobacco-related DNA methylation signatures. They belong to 95 genes which are involved in hsa04512, hsa04151, and other important pathways. At gene regulation level, 33 genes are uncovered to be highly related to signature probes by both BIMC and SRC methods. Among them, FARSB and other eight genes were uncovered as Hub genes in the gene regulatory network. Meanwhile, the PPI network about these 33 genes showed that MAGOH, FYN, and other five genes were the most connected core genes among them. These analysis results may provide clues for a clear biological interpretation in the molecular mechanism of tumorigenesis. Moreover, the identified signature probes may serve as potential drug targets for the precision medicine of LUAD.
Collapse
|
53
|
Pittman GS, Wang X, Campbell MR, Coulter SJ, Olson JR, Pavuk M, Birnbaum LS, Bell DA. Polychlorinated biphenyl exposure and DNA methylation in the Anniston Community Health Survey. Epigenetics 2020; 15:337-357. [PMID: 31607210 PMCID: PMC7153539 DOI: 10.1080/15592294.2019.1666654] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/26/2019] [Accepted: 09/06/2019] [Indexed: 12/12/2022] Open
Abstract
Anniston, Alabama was home to a major polychlorinated biphenyl (PCB) production facility from 1929 until 1971. The Anniston Community Health Survey I and II (ACHS-I 2005-2007, ACHS-II 2013-2014) were conducted to explore the effects of PCB exposures. In this report we examined associations between PCB exposure and DNA methylation in whole blood using EPIC arrays (ACHS-I, n = 518; ACHS-II, n = 299). For both cohorts, 35 PCBs were measured in serum. We modelled methylation versus PCB wet-weight concentrations for: the sum of 35 PCBs, mono-ortho substituted PCBs, di-ortho substituted PCBs, tri/tetra-ortho substituted PCBs, oestrogenic PCBs, and antiestrogenic PCBs. Using robust multivariable linear regression, we adjusted for age, race, sex, smoking, total lipids, and six blood cell-type percentages. We carried out a two-stage analysis; discovery in ACHS-I followed by replication in ACHS-II. In ACHS-I, we identified 28 associations (17 unique CpGs) at p ≤ 6.70E-08 and 369 associations (286 unique CpGs) at FDR p ≤ 5.00E-02. A large proportion of the genes have been observed to interact with PCBs or dioxins in model studies. Among the 28 genome-wide significant CpG/PCB associations, 14 displayed replicated directional effects in ACHS-II; however, only one in ACHS-II was statistically significant at p ≤ 1.70E-04. While we identified many novel CpGs significantly associated with PCB exposures in ACHS-I, the differential methylation was modest and the effect was attenuated seven years later in ACHS-II, suggesting a lack of persistence of the associations between PCB exposures and altered DNA methylation in blood cells.
Collapse
Affiliation(s)
- Gary S. Pittman
- National Institute of Environmental Health Sciences, RTP, Durham, NC, USA
| | - Xuting Wang
- National Institute of Environmental Health Sciences, RTP, Durham, NC, USA
| | | | | | | | - Marian Pavuk
- ATSDR, Center for Disease Control, Atlanta, Fulton, GA, USA
| | | | - Douglas A. Bell
- National Institute of Environmental Health Sciences, RTP, Durham, NC, USA
| |
Collapse
|
54
|
Lei MK, Gibbons FX, Simons RL, Philibert RA, Beach SRH. The Effect of Tobacco Smoking Differs across Indices of DNA Methylation-Based Aging in an African American Sample: DNA Methylation-Based Indices of Smoking Capture These Effects. Genes (Basel) 2020; 11:E311. [PMID: 32183340 PMCID: PMC7140795 DOI: 10.3390/genes11030311] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/09/2020] [Accepted: 03/12/2020] [Indexed: 01/09/2023] Open
Abstract
Smoking is one of the leading preventable causes of morbidity and mortality worldwide, prompting interest in its association with DNA methylation-based measures of biological aging. Considerable progress has been made in developing DNA methylation-based measures that correspond to self-reported smoking status. In addition, assessment of DNA methylation-based aging has been expanded to better capture individual differences in risk for morbidity and mortality. Untested to date, however, is whether smoking is similarly related to older and newer indices of DNA methylation-based aging, and whether DNA methylation-based indices of smoking can be used in lieu of self-reported smoking to examine effects on DNA methylation-based aging measures. In the current investigation we examine mediation of the impact of self-reported cigarette consumption on accelerated, intrinsic DNA methylation-based aging using indices designed to predict chronological aging, phenotypic aging, and mortality risk, as well as a newly developed DNA methylation-based measure of telomere length. Using a sample of 500 African American middle aged smokers and non-smokers, we found that a) self-reported cigarette consumption was associated with accelerated intrinsic DNA methylation-based aging on some but not all DNA methylation-based aging indices, b) for those aging outcomes associated with self-reported cigarette consumption, DNA methylation-based indicators of smoking typically accounted for greater variance than did self-reported cigarette consumption, and c) self-reported cigarette consumption effects on DNA methylation-based aging indices typically were fully mediated by DNA methylation-based indicators of smoking (e.g., PACKYRS from GrimAge; or cg05575921 CpG site). Results suggest that when DNA methylation-based indices of smoking are substituted for self-reported assessments of smoking, they will typically fully reflect the varied impact of cigarette smoking on intrinsic, accelerated DNA methylation-based aging.
Collapse
Affiliation(s)
- Man-Kit Lei
- Department of Sociology, University of Georgia, Athens, GA 30602, USA; (M.-K.L.); (R.L.S.)
| | - Frederick X. Gibbons
- Department of Psychological Sciences, University of Connecticut, Storrs, CT 06269, USA;
| | - Ronald L. Simons
- Department of Sociology, University of Georgia, Athens, GA 30602, USA; (M.-K.L.); (R.L.S.)
| | - Robert A. Philibert
- Department of Psychiatry, University of Iowa, Iowa, IA 52242, USA;
- Behavioral Diagnostics, Coralville, Iowa, IA 52241, USA
| | - Steven R. H. Beach
- Department of Psychology, University of Georgia, Athens, GA 30602, USA
- Center for Family Research, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
55
|
Machnik M, Oleksiewicz U. Dynamic Signatures of the Epigenome: Friend or Foe? Cells 2020; 9:cells9030653. [PMID: 32156057 PMCID: PMC7140607 DOI: 10.3390/cells9030653] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/24/2020] [Accepted: 03/04/2020] [Indexed: 12/12/2022] Open
Abstract
Highly dynamic epigenetic signaling is influenced mainly by (micro)environmental stimuli and genetic factors. The exact mechanisms affecting particular epigenomic patterns differ dependently on the context. In the current review, we focus on the causes and effects of the dynamic signatures of the human epigenome as evaluated with the high-throughput profiling data and single-gene approaches. We will discuss three different aspects of phenotypic outcomes occurring as a consequence of epigenetics interplaying with genotype and environment. The first issue is related to the cases of environmental impacts on epigenetic profile, and its adverse and advantageous effects related to human health and evolutionary adaptation. The next topic will present a model of the interwoven co-evolution of genetic and epigenetic patterns exemplified with transposable elements (TEs) and their epigenetic repressors Krüppel-associated box zinc finger proteins (KRAB–ZNFs). The third aspect concentrates on the mitosis-based microevolution that takes place during carcinogenesis, leading to clonal diversity and expansion of tumor cells. The whole picture of epigenome plasticity and its role in distinct biological processes is still incomplete. However, accumulating data define epigenomic dynamics as an essential co-factor driving adaptation at the cellular and inter-species levels with a benefit or disadvantage to the host.
Collapse
Affiliation(s)
- Marta Machnik
- Department of Cancer Immunology, Poznan University of Medical Sciences, 60-806 Poznan, Poland;
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Urszula Oleksiewicz
- Department of Cancer Immunology, Poznan University of Medical Sciences, 60-806 Poznan, Poland;
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
- Correspondence:
| |
Collapse
|
56
|
Philibert R, Dogan M, Beach SRH, Mills JA, Long JD. AHRR methylation predicts smoking status and smoking intensity in both saliva and blood DNA. Am J Med Genet B Neuropsychiatr Genet 2020; 183:51-60. [PMID: 31456352 DOI: 10.1002/ajmg.b.32760] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/15/2019] [Accepted: 08/12/2019] [Indexed: 01/21/2023]
Abstract
Many existing DNA repositories do not have robust characterizations of smoking, while for many currently ongoing studies, the advent of vaping has rendered traditional cotinine-based methods of determining smoking status unreliable. Previously, we have shown that methylation status at cg05575921 in whole blood DNA can reliably predict cigarette consumption. However, whether methylation status in saliva can be used similarly has yet to be established. Herein, we use DNA from 418 biochemically confirmed smokers or nonsmokers to compare and contrast the utility of cg05575921 in classifying and quantifying cigarette smoking. Using whole blood DNA, a model incorporating age, gender, and methylation status had a receiver operating characteristic (ROC) area under the curve (AUC) for predicting smoking status of 0.995 with a nonlinear demethylation response to smoking. Using saliva DNA, the ROC AUC for predicting smoking was 0.971 with the plot of the relationship of DNA methylation to daily cigarette consumption being very similar to that seen for whole blood DNA. The addition of information from another methylation marker designed to correct for cellular heterogeneity improved the AUC for saliva DNA to 0.981. Finally, in 31 subjects who reported quitting smoking 10 or more years previously, cg05575921 methylation was nonsignificantly different from controls. We conclude that DNA methylation status at cg05575921 in DNA from whole blood or saliva predicts smoking status and daily cigarette consumption. We suggest these epigenetic assessments for objectively ascertaining smoking status will find utility in research, clinical, and civil applications.
Collapse
Affiliation(s)
- Robert Philibert
- Department of Psychiatry, University of Iowa, Iowa City, Iowa.,Behavioral Diagnostics LLC, Coralville, Iowa
| | - Meeshanthini Dogan
- Department of Psychiatry, University of Iowa, Iowa City, Iowa.,Cardio Diagnostics LLC, Coralville, Iowa
| | - Steven R H Beach
- Center for Family Studies, University of Georgia, Athens, Georgia
| | - James A Mills
- Department of Psychiatry, University of Iowa, Iowa City, Iowa
| | - Jeffrey D Long
- Department of Psychiatry, University of Iowa, Iowa City, Iowa.,Department of Biostatistics, University of Iowa, Iowa City, Iowa
| |
Collapse
|
57
|
McCord B, Gauthier Q, Alghanim H, Antunes J, Fernandez Tejero N, Duncan G, Balamurugan K. Applications of epigenetic methylation in body fluid identification, age determination and phenotyping. FORENSIC SCIENCE INTERNATIONAL GENETICS SUPPLEMENT SERIES 2019. [DOI: 10.1016/j.fsigss.2019.10.061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
58
|
Corley J, Cox SR, Harris SE, Hernandez MV, Maniega SM, Bastin ME, Wardlaw JM, Starr JM, Marioni RE, Deary IJ. Epigenetic signatures of smoking associate with cognitive function, brain structure, and mental and physical health outcomes in the Lothian Birth Cohort 1936. Transl Psychiatry 2019; 9:248. [PMID: 31591380 PMCID: PMC6779733 DOI: 10.1038/s41398-019-0576-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 06/29/2019] [Indexed: 12/18/2022] Open
Abstract
Recent advances in genome-wide DNA methylation (DNAm) profiling for smoking behaviour have given rise to a new, molecular biomarker of smoking exposure. It is unclear whether a smoking-associated DNAm (epigenetic) score has predictive value for ageing-related health outcomes which is independent of contributions from self-reported (phenotypic) smoking measures. Blood DNA methylation levels were measured in 895 adults aged 70 years in the Lothian Birth Cohort 1936 (LBC1936) study using the Illumina 450K assay. A DNA methylation score based on 230 CpGs was used as a proxy for smoking exposure. Associations between smoking variables and health outcomes at age 70 were modelled using general linear modelling (ANCOVA) and logistic regression. Additional analyses of smoking with brain MRI measures at age 73 (n = 532) were performed. Smoking-DNAm scores were positively associated with self-reported smoking status (P < 0.001, eta-squared ɳ2 = 0.63) and smoking pack years (r = 0.69, P < 0.001). Higher smoking DNAm scores were associated with variables related to poorer cognitive function, structural brain integrity, physical health, and psychosocial health. Compared with phenotypic smoking, the methylation marker provided stronger associations with all of the cognitive function scores, especially visuospatial ability (P < 0.001, partial eta-squared ɳp2 = 0.022) and processing speed (P < 0.001, ɳp2 = 0.030); inflammatory markers (all P < 0.001, ranges from ɳp2 = 0.021 to 0.030); dietary patterns (healthy diet (P < 0.001, ɳp2 = 0.052) and traditional diet (P < 0.001, ɳp2 = 0.032); stroke (P = 0.006, OR 1.48, 95% CI 1.12, 1.96); mortality (P < 0.001, OR 1.59, 95% CI 1.42, 1.79), and at age 73; with MRI volumetric measures (all P < 0.001, ranges from ɳp2 = 0.030 to 0.052). Additionally, education was the most important life-course predictor of lifetime smoking tested. Our results suggest that a smoking-associated methylation biomarker typically explains a greater proportion of the variance in some smoking-related morbidities in older adults, than phenotypic measures of smoking exposure, with some of the accounted-for variance being independent of phenotypic smoking status.
Collapse
Affiliation(s)
- Janie Corley
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK.
| | - Simon R Cox
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Maria Valdéz Hernandez
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Brain Research Imaging Centre, Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Susana Muñoz Maniega
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Brain Research Imaging Centre, Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Mark E Bastin
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Brain Research Imaging Centre, Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Joanna M Wardlaw
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Brain Research Imaging Centre, Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Royal Victoria Building, Western General Hospital, Porterfield Road, Edinburgh, UK
| | - Riccardo E Marioni
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| |
Collapse
|
59
|
Kuniyoshi KM, Rehan VK. The impact of perinatal nicotine exposure on fetal lung development and subsequent respiratory morbidity. Birth Defects Res 2019; 111:1270-1283. [PMID: 31580538 DOI: 10.1002/bdr2.1595] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/13/2019] [Accepted: 09/05/2019] [Indexed: 01/18/2023]
Abstract
Maternal smoking during pregnancy remains as a significant public health crisis as it did decades ago. Although its prevalence is decreasing in high-income countries, it has worsened globally, along with a concerning emergence of electronic-cigarette usage within the last two decades. Extensive epidemiologic and experimental evidence exists from both human and animal studies, demonstrating the detrimental long-term pulmonary outcomes in the offspring of mothers who smoke during pregnancy. Even secondhand and thirdhand smoke exposure to the developing lung might be as or even more harmful than firsthand smoke exposure. Furthermore, these effects are not limited only to the exposed progeny, but can also be transmitted transgenerationally. There is compelling evidence to support that the majority of the effects of perinatal smoke exposure on the developing lung, including the transgenerational transmission of asthma, is mediated by nicotine. Nicotine exposure induces cell-specific molecular changes in lungs, which offers a unique opportunity to prevent, halt, and/or reverse the resultant damage through targeted molecular interventions. Experimentally, the proposed interventions, such as administration of peroxisome proliferator-activated receptor gamma (PPARγ) agonists can not only block but also potentially reverse the perinatal nicotine exposure-induced respiratory morbidity in the exposed offspring. However, the development of a safe and effective intervention is still many years away. In the meantime, electropuncture at specific acupoints appears to be emerging as a more practical and safe physiologic approach to block the harmful pulmonary consequences of perinatal nicotine exposure.
Collapse
Affiliation(s)
- Katherine M Kuniyoshi
- Department of Pediatrics, David Geffen School of Medicine, The Lundquist Institute for Biomedical Innovation at Harbor, UCLA Medical Center, Torrance, California
| | - Virender K Rehan
- Department of Pediatrics, David Geffen School of Medicine, The Lundquist Institute for Biomedical Innovation at Harbor, UCLA Medical Center, Torrance, California
| |
Collapse
|
60
|
Kaur G, Begum R, Thota S, Batra S. A systematic review of smoking-related epigenetic alterations. Arch Toxicol 2019; 93:2715-2740. [PMID: 31555878 DOI: 10.1007/s00204-019-02562-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/02/2019] [Indexed: 02/06/2023]
Abstract
The aim of this study is to provide a systematic review of the known epigenetic alterations caused by cigarette smoke; establish an evidence-based perspective of their clinical value for screening, diagnosis, and treatment of smoke-related disorders; and discuss the challenges and ethical concerns associated with epigenetic studies. A well-defined, reproducible search strategy was employed to identify relevant literature (clinical, cellular, and animal-based) between 2000 and 2019 based on AMSTAR guidelines. A total of 80 studies were identified that reported alterations in DNA methylation, histone modifications, and miRNA expression following exposure to cigarette smoke. Changes in DNA methylation were most extensively documented for genes including AHRR, F2RL3, DAPK, and p16 after exposure to cigarette smoke. Likewise, miR16, miR21, miR146, and miR222 were identified to be differentially expressed in smokers and exhibit potential as biomarkers for determining susceptibility to COPD. We also identified 22 studies highlighting the transgenerational effects of maternal and paternal smoking on offspring. This systematic review lists the epigenetic events/alterations known to occur in response to cigarette smoke exposure and identifies the major genes and miRNAs that are potential targets for translational research in associated pathologies. Importantly, the limitations and ethical concerns related to epigenetic studies are also highlighted, as are the effects on the ability to address specific questions associated with exposure to tobacco/cigarette smoke. In the future, improved interpretation of epigenetic signatures will lead to their increased use as biomarkers and/or in drug development.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Laboratory of Pulmonary Immuno-toxicology, Department of Environmental Toxicology, 129 Health Research Centre, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Rizwana Begum
- Laboratory of Pulmonary Immuno-toxicology, Department of Environmental Toxicology, 129 Health Research Centre, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Shilpa Thota
- Laboratory of Pulmonary Immuno-toxicology, Department of Environmental Toxicology, 129 Health Research Centre, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Sanjay Batra
- Laboratory of Pulmonary Immuno-toxicology, Department of Environmental Toxicology, 129 Health Research Centre, Southern University and A&M College, Baton Rouge, LA, 70813, USA.
| |
Collapse
|
61
|
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] [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
|
62
|
Ding Y, Li Q, Feng Q, Xu D, Wu C, Zhao J, Zhou X, Yang Y, Niu H, He P, Xing L. CYP2B6 genetic polymorphisms influence chronic obstructive pulmonary disease susceptibility in the Hainan population. Int J Chron Obstruct Pulmon Dis 2019; 14:2103-2115. [PMID: 31564857 PMCID: PMC6733340 DOI: 10.2147/copd.s214961] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/23/2019] [Indexed: 11/23/2022] Open
Abstract
Introduction Chronic obstructive pulmonary disease (COPD) is a lung disease closely related to exposure to exogenous substances. CYP2B6 can activate many exogenous substances, which in turn affect lung cells. The aim of this study was to assess the association of single-nucleotide polymorphisms (SNPs) in CYP2B6 with COPD risk in a Chinese Han population. Materials and methods Genotypes of the five candidate SNPs in CYP2B6 were identified among 318 cases and 508 healthy controls with an Agena MassARRAY method. The association between CYP2B6 polymorphisms and COPD risk was evaluated using genetic models and haplotype analyses. Results In allele model, we observed that rs4803420 G and rs1038376 A were related to COPD risk. And rs4803420 G/T and G/T-T/T were related to a decreased COPD risk compared to GG genotype in the co-dominant and dominant models, respectively. When comparing with the AA genotype, rs1038376 A/T and A/T-T/T were associated with an increased COPD risk in the co-dominant and dominant models, respectively. Further gender stratification co-dominant and dominant models analysis showed that genotype G/T and G/T-T/T of rs4803420, and genotype A/T and A/T-T/T of rs1038376 were significantly associated with COPD risk compared to the wide type in males and females, while allele C of rs12979270 was only associated with COPD risk in females. Smoking status stratification analysis showed that rs12979270 C was significantly associated with an increased COPD risk under the allele model compared with allele A in the smoking subgroup. Haplotype analysis showed that haplotype GTA and TAA were related to COPD risk. Conclusion Our data is the first to demonstrate that CYP2B6 polymorphisms may exert effects on COPD susceptibility in the Chinese Han population.
Collapse
Affiliation(s)
- Yipeng Ding
- Department of General Practice, Hainan General Hospital, Haikou, Hainan570311, People’s Republic of China
| | - Quanni Li
- Department of General Practice, Hainan General Hospital, Haikou, Hainan570311, People’s Republic of China
| | - Qiong Feng
- Hainan General Hospital, University of South China, Haikou, Hainan570311, People’s Republic of China
| | - Dongchuan Xu
- Department of General Practice, Hainan General Hospital, Haikou, Hainan570311, People’s Republic of China
| | - Cibing Wu
- Hainan General Hospital, University of South China, Haikou, Hainan570311, People’s Republic of China
| | - Jie Zhao
- Hainan General Hospital, University of South China, Haikou, Hainan570311, People’s Republic of China
| | - Xiaoli Zhou
- Department of General Practice, Hainan General Hospital, Haikou, Hainan570311, People’s Republic of China
| | - Yixiu Yang
- Department of General Practice, Hainan General Hospital, Haikou, Hainan570311, People’s Republic of China
| | - Huan Niu
- Department of General Practice, Hainan General Hospital, Haikou, Hainan570311, People’s Republic of China
| | - Ping He
- Department of General Practice, Hainan General Hospital, Haikou, Hainan570311, People’s Republic of China
| | - Lihua Xing
- Department of Respiratory Intensive Care Unit (RICU), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan45000, People’s Republic of China
| |
Collapse
|
63
|
Richter GM, Kruppa J, Munz M, Wiehe R, Häsler R, Franke A, Martins O, Jockel-Schneider Y, Bruckmann C, Dommisch H, Schaefer AS. A combined epigenome- and transcriptome-wide association study of the oral masticatory mucosa assigns CYP1B1 a central role for epithelial health in smokers. Clin Epigenetics 2019; 11:105. [PMID: 31331382 PMCID: PMC6647091 DOI: 10.1186/s13148-019-0697-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/18/2019] [Indexed: 01/08/2023] Open
Abstract
Background The oral mucosa has an important role in maintaining barrier integrity at the gateway to the gastrointestinal and respiratory tracts. Smoking is a strong environmental risk factor for the common oral inflammatory disease periodontitis and oral cancer. Cigarette smoke affects gene methylation and expression in various tissues. This is the first epigenome-wide association study (EWAS) that aimed to identify biologically active methylation marks of the oral masticatory mucosa that are associated with smoking. Results Ex vivo biopsies of 18 current smokers and 21 never smokers were analysed with the Infinium Methylation EPICBeadChip and combined with whole transcriptome RNA sequencing (RNA-Seq; 16 mio reads per sample) of the same samples. We analysed the associations of CpG methylation values with cigarette smoking and smoke pack year (SPY) levels in an analysis of covariance (ANCOVA). Nine CpGs were significantly associated with smoking status, with three CpGs mapping to the genetic region of CYP1B1 (cytochrome P450 family 1 subfamily B member 1; best p = 5.5 × 10−8) and two mapping to AHRR (aryl-hydrocarbon receptor repressor; best p = 5.9 × 10−9). In the SPY analysis, 61 CpG sites at 52 loci showed significant associations of the quantity of smoking with changes in methylation values. Here, the most significant association located to the gene CYP1B1, with p = 4.0 × 10−10. RNA-Seq data showed significantly increased expression of CYP1B1 in smokers compared to non-smokers (p = 2.2 × 10−14), together with 13 significantly upregulated transcripts. Six transcripts were significantly downregulated. No differential expression was observed for AHRR. In vitro studies with gingival fibroblasts showed that cigarette smoke extract directly upregulated the expression of CYP1B1. Conclusion This study validated the established role of CYP1B1 and AHRR in xenobiotic metabolism of tobacco smoke and highlights the importance of epigenetic regulation for these genes. For the first time, we give evidence of this role for the oral masticatory mucosa. Electronic supplementary material The online version of this article (10.1186/s13148-019-0697-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gesa M Richter
- Department of Periodontology and Synoptic Dentistry, Institute for Dental and Craniofacial Sciences, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Aßmannshauser Str. 4-6, 14197, Berlin, Germany.
| | - Jochen Kruppa
- Institute for Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Matthias Munz
- Department of Periodontology and Synoptic Dentistry, Institute for Dental and Craniofacial Sciences, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Aßmannshauser Str. 4-6, 14197, Berlin, Germany.,Medical Systems Biology Group, Institute of Experimental Dermatology, Institute for Cardiogenetics, University of Lübeck, 23562, Lübeck, Germany
| | - Ricarda Wiehe
- Department of Periodontology and Synoptic Dentistry, Institute for Dental and Craniofacial Sciences, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Aßmannshauser Str. 4-6, 14197, Berlin, Germany
| | - Robert Häsler
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany
| | - Orlando Martins
- Institute of Periodontology, Dentistry Department, Faculty of Medicine, University of Coimbra, Av. Bissaya Barreto, Bloco de Celas, 3000-075, Coimbra, Portugal
| | - Yvonne Jockel-Schneider
- Department of Periodontology, Clinic of Preventive Dentistry and Periodontology, University Medical Center of the Julius-Maximilians-University, Pleicherwall, 97070, Würzburg, Germany
| | - Corinna Bruckmann
- Department of Conservative Dentistry and Periodontology, Medical University Vienna, School of Dentistry, Sensengasse 2a, 1090, Vienna, Austria
| | - Henrik Dommisch
- Department of Periodontology and Synoptic Dentistry, Institute for Dental and Craniofacial Sciences, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Aßmannshauser Str. 4-6, 14197, Berlin, Germany
| | - Arne S Schaefer
- Department of Periodontology and Synoptic Dentistry, Institute for Dental and Craniofacial Sciences, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Aßmannshauser Str. 4-6, 14197, Berlin, Germany
| |
Collapse
|
64
|
Ringh MV, Hagemann-Jensen M, Needhamsen M, Kular L, Breeze CE, Sjöholm LK, Slavec L, Kullberg S, Wahlström J, Grunewald J, Brynedal B, Liu Y, Almgren M, Jagodic M, Öckinger J, Ekström TJ. Tobacco smoking induces changes in true DNA methylation, hydroxymethylation and gene expression in bronchoalveolar lavage cells. EBioMedicine 2019; 46:290-304. [PMID: 31303497 PMCID: PMC6710853 DOI: 10.1016/j.ebiom.2019.07.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 12/21/2022] Open
Abstract
Background While smoking is known to associate with development of multiple diseases, the underlying mechanisms are still poorly understood. Tobacco smoking can modify the chemical integrity of DNA leading to changes in transcriptional activity, partly through an altered epigenetic state. We aimed to investigate the impact of smoking on lung cells collected from bronchoalveolar lavage (BAL). Methods We profiled changes in DNA methylation (5mC) and its oxidised form hydroxymethylation (5hmC) using conventional bisulphite (BS) treatment and oxidative bisulphite treatment with Illumina Infinium MethylationEPIC BeadChip, and examined gene expression by RNA-seq in healthy smokers. Findings We identified 1667 total 5mC + 5hmC, 1756 5mC and 67 5hmC differentially methylated positions (DMPs) between smokers and non-smokers (FDR-adjusted P <.05, absolute Δβ >0.15). Both 5mC DMPs and to a lesser extent 5mC + 5hmC were predominantly hypomethylated. In contrast, almost all 5hmC DMPs were hypermethylated, supporting the hypothesis that smoking-associated oxidative stress can lead to DNA demethylation, via the established sequential oxidation of which 5hmC is the first step. While we confirmed differential methylation of previously reported smoking-associated 5mC + 5hmC CpGs using former generations of BeadChips in alveolar macrophages, the large majority of identified DMPs, 5mC + 5hmC (1639/1667), 5mC (1738/1756), and 5hmC (67/67), have not been previously reported. Most of these novel smoking-associating sites are specific to the EPIC BeadChip and, interestingly, many of them are associated to FANTOM5 enhancers. Transcriptional changes affecting 633 transcripts were consistent with DNA methylation profiles and converged to alteration of genes involved in migration, signalling and inflammatory response of immune cells. Interpretation Collectively, these findings suggest that tobacco smoke exposure epigenetically modifies BAL cells, possibly involving a continuous active demethylation and subsequent increased activity of inflammatory processes in the lungs. Fund The study was supported by the Swedish Research Council, the Swedish Heart-Lung Foundation, the Stockholm County Council (ALF), the King Gustav's and Queen Victoria's Freemasons' Foundation, Knut and Alice Wallenberg Foundation, Neuro Sweden, and the Swedish MS foundation.
Collapse
Affiliation(s)
- Mikael V Ringh
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden.
| | - Michael Hagemann-Jensen
- Department of Medicine, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
| | - Maria Needhamsen
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
| | - Lara Kular
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
| | - Charles E Breeze
- Altius Institute for Biomedical Sciences, Seattle, USA; UCL Cancer Institute, University College London, London, United Kingdom
| | - Louise K Sjöholm
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
| | - Lara Slavec
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
| | - Susanna Kullberg
- Department of Medicine, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden; Department of Respiratory Medicine, Theme Inflammation and Infection, Karolinska University Hospital, Stockholm, Sweden
| | - Jan Wahlström
- Department of Medicine, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
| | - Johan Grunewald
- Department of Medicine, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
| | - Boel Brynedal
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yun Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Malin Almgren
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
| | - Johan Öckinger
- Department of Medicine, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
| | - Tomas J Ekström
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden.
| |
Collapse
|
65
|
Imboden M, Wielscher M, Rezwan FI, Amaral A, Schaffner E, Jeong A, Beckmeyer-Borowko A, Harris SE, Starr JM, Deary I, Flexeder C, Waldenberger M, Peters A, Schulz H, Chen S, Sunny SK, Karmaus WJ, Jiang Y, Erhart G, Kronenberg F, Arathimos R, Sharp GC, Henderson AJ, Fu Y, Piirilä P, Pietiläinen KH, Ollikainen M, Johansson A, Gyllensten U, de Vries M, van der Plaat DA, de Jong K, Boezen HM, Hall I, Tobin MD, Jarvelin MR, Holloway J, Jarvis D, Probst-Hensch NM. Epigenome-wide association study of lung function level and its change. Eur Respir J 2019; 54:13993003.00457-2019. [PMID: 31073081 PMCID: PMC6610463 DOI: 10.1183/13993003.00457-2019] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 03/27/2019] [Indexed: 12/19/2022]
Abstract
Previous reports link differential DNA methylation (DNAme) to environmental exposures that are associated with lung function. Direct evidence on lung function DNAme is, however, limited. We undertook an agnostic epigenome-wide association study (EWAS) on pre-bronchodilation lung function and its change in adults.In a discovery-replication EWAS design, DNAme in blood and spirometry were measured twice, 6-15 years apart, in the same participants of three adult population-based discovery cohorts (n=2043). Associated DNAme markers (p<5×10-7) were tested in seven replication cohorts (adult: n=3327; childhood: n=420). Technical bias-adjusted residuals of a regression of the normalised absolute β-values on control probe-derived principle components were regressed on level and change of forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC) and their ratio (FEV1/FVC) in the covariate-adjusted discovery EWAS. Inverse-variance-weighted meta-analyses were performed on results from discovery and replication samples in all participants and never-smokers.EWAS signals were enriched for smoking-related DNAme. We replicated 57 lung function DNAme markers in adult, but not childhood samples, all previously associated with smoking. Markers not previously associated with smoking failed replication. cg05575921 (AHRR (aryl hydrocarbon receptor repressor)) showed the statistically most significant association with cross-sectional lung function (FEV1/FVC: pdiscovery=3.96×10-21 and pcombined=7.22×10-50). A score combining 10 DNAme markers previously reported to mediate the effect of smoking on lung function was associated with lung function (FEV1/FVC: p=2.65×10-20).Our results reveal that lung function-associated methylation signals in adults are predominantly smoking related, and possibly of clinical utility in identifying poor lung function and accelerated decline. Larger studies with more repeat time-points are needed to identify lung function DNAme in never-smokers and in children.
Collapse
Affiliation(s)
- Medea Imboden
- Chronic Disease Epidemiology Unit, Dept of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland,University of Basel, Basel, Switzerland,These authors contributed equally to this work
| | - Matthias Wielscher
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK,Dept of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK,These authors contributed equally to this work
| | - Faisal I. Rezwan
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK,These authors contributed equally to this work
| | - André F.S. Amaral
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK,Population Health and Occupational Disease, NHLI, Imperial College London, London, UK,These authors contributed equally to this work
| | - Emmanuel Schaffner
- Chronic Disease Epidemiology Unit, Dept of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland,University of Basel, Basel, Switzerland
| | - Ayoung Jeong
- Chronic Disease Epidemiology Unit, Dept of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland,University of Basel, Basel, Switzerland
| | - Anna Beckmeyer-Borowko
- Chronic Disease Epidemiology Unit, Dept of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland,University of Basel, Basel, Switzerland
| | - Sarah E. Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK,Medical Genetics Section, University of Edinburgh Centre for Genomic and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, UK
| | - John M. Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK,Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - Ian J. Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK,Dept of Psychology, University of Edinburgh, Edinburgh, UK
| | - Claudia Flexeder
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Melanie Waldenberger
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany,Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany,Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Holger Schulz
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany,Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Su Chen
- Dept of Mathematical Sciences, University of Memphis, Memphis, TN, USA
| | - Shadia Khan Sunny
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Wilfried J.J. Karmaus
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Yu Jiang
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Gertraud Erhart
- Division of Genetic Epidemiology, Dept of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Dept of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ryan Arathimos
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK,Dept of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Gemma C. Sharp
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK,Dept of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK,Bristol Dental School, University of Bristol, Bristol, UK
| | | | - Yu Fu
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Päivi Piirilä
- Unit of Clinical Physiology, HUS Medical Imaging Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Kirsi H. Pietiläinen
- Obesity Research Unit, Research Programs Unit, University of Helsinki, Helsinki, Finland,Abdominal Center, Endocrinology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Asa Johansson
- Dept of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ulf Gyllensten
- Dept of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Maaike de Vries
- University of Groningen, University Medical Center Groningen, Dept of Epidemiology, Groningen, The Netherlands,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Diana A. van der Plaat
- University of Groningen, University Medical Center Groningen, Dept of Epidemiology, Groningen, The Netherlands,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Kim de Jong
- University of Groningen, University Medical Center Groningen, Dept of Epidemiology, Groningen, The Netherlands,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H. Marike Boezen
- University of Groningen, University Medical Center Groningen, Dept of Epidemiology, Groningen, The Netherlands,Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ian P. Hall
- Division of Respiratory Medicine, University of Nottingham, Nottingham, UK,National Institute for Health Research, Nottingham Biomedical Research Centre, Nottingham University Hospitals, Nottingham, UK
| | - Martin D. Tobin
- Dept of Health Sciences, University of Leicester, Leicester, UK,National Institute of Health Research Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Marjo-Riitta Jarvelin
- Dept of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK,Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland,Biocenter Oulu, University of Oulu, Oulu, Finland,Unit of Primary Health Care, Oulu University Hospital, Oulu, Finland,Dept of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK,These authors contributed equally to this work
| | - John W. Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK,These authors contributed equally to this work
| | - Deborah Jarvis
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK,Population Health and Occupational Disease, NHLI, Imperial College London, London, UK,These authors contributed equally to this work
| | - Nicole M. Probst-Hensch
- Chronic Disease Epidemiology Unit, Dept of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland,University of Basel, Basel, Switzerland,These authors contributed equally to this work
| |
Collapse
|
66
|
London SJ. Methylation, smoking, and reduced lung function. Eur Respir J 2019; 54:54/1/1900920. [PMID: 31273037 DOI: 10.1183/13993003.00920-2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 01/04/2023]
Affiliation(s)
- Stephanie J London
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Dept of Health and Human Services, Research Triangle Park, NC, USA
| |
Collapse
|
67
|
Smoking-Related DNA Methylation is Associated with DNA Methylation Phenotypic Age Acceleration: The Veterans Affairs Normative Aging Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16132356. [PMID: 31277270 PMCID: PMC6651499 DOI: 10.3390/ijerph16132356] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/29/2019] [Accepted: 07/01/2019] [Indexed: 12/17/2022]
Abstract
DNA methylation may play a critical role in aging and age-related diseases. DNA methylation phenotypic age (DNAmPhenoAge) is a new aging biomarker and predictor of chronic disease risk. While smoking is a strong risk factor for chronic diseases and influences methylation, its influence on DNAmPhenoAge is unknown. We investigated associations of self-reported and epigenetic smoking indicators with DNAmPhenoAge acceleration in a longitudinal aging study in eastern Massachusetts. DNA methylation was measured in whole blood samples from multiple visits for 692 male participants in the Veterans Affairs Normative Aging Study during 1999–2013. Acceleration was defined using residuals from linear regression of the DNAmPhenoAge on the chronological age. Cumulative smoking (pack-years) was significantly associated with DNAmPhenoAge acceleration, whereas self-reported smoking status was not. We observed significant validated associations between smoking-related loci and DNAmPhenoAge acceleration for 52 CpG sites, where 18 were hypomethylated and 34 were hypermethylated, mapped to 16 genes. The AHRR gene had the most loci (N = 8) among the 16 genes. We generated a smoking aging index based on these 52 loci, which showed positive significant associations with DNAmPhenoAge acceleration. These epigenetic biomarkers may help to predict age-related risks driven by smoking.
Collapse
|
68
|
Affiliation(s)
- Christine Beedham
- Honorary Senior Lecturer, Faculty of Life Sciences, School of Pharmacy and Medical Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
69
|
Freudenheim JL, Shields PG, Song MA, Smiraglia D. DNA Methylation and Smoking: Implications for Understanding Effects of Electronic Cigarettes. CURR EPIDEMIOL REP 2019. [DOI: 10.1007/s40471-019-00191-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
70
|
Fragou D, Pakkidi E, Aschner M, Samanidou V, Kovatsi L. Smoking and DNA methylation: Correlation of methylation with smoking behavior and association with diseases and fetus development following prenatal exposure. Food Chem Toxicol 2019; 129:312-327. [PMID: 31063835 DOI: 10.1016/j.fct.2019.04.059] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 12/13/2022]
Abstract
Among epigenetic mechanisms, DNA methylation has been widely studied with respect to many environmental factors. Smoking is a common factor which affects both global and gene-specific DNA methylation. It is supported that smoking directly affects DNA methylation, and these effects contribute to the development and progression of various diseases, such as cancer, lung and cardiovascular diseases and male infertility. In addition, prenatal smoking influences the normal development of the fetus via DNA methylation changes. The DNA methylation profile and its smoking-induced alterations helps to distinguish current from former smokers and non-smokers and can be used to predict the risk for the development of a disease. This review summarizes the DNA methylation changes induced by smoking, their correlation with smoking behavior and their association with various diseases and fetus development.
Collapse
Affiliation(s)
- Domniki Fragou
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Greece
| | - Eleni Pakkidi
- Laboratory of Analytical Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, Greece
| | - Michael Aschner
- Departments of Molecular Pharmacology, Neuroscience, and Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Victoria Samanidou
- Laboratory of Analytical Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, Greece
| | - Leda Kovatsi
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Greece.
| |
Collapse
|
71
|
Su KY, Li MC, Lee NW, Ho BC, Cheng CL, Chuang YC, Yu SL, Guo YL. Perinatal polychlorinated biphenyls and polychlorinated dibenzofurans exposure are associated with DNA methylation changes lasting to early adulthood: Findings from Yucheng second generation. ENVIRONMENTAL RESEARCH 2019; 170:481-486. [PMID: 30640082 DOI: 10.1016/j.envres.2019.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 11/29/2018] [Accepted: 01/02/2019] [Indexed: 05/17/2023]
Abstract
Epigenome-wide DNA methylation has not been studied in men perinatally exposed to PCBs and dioxins. Therefore, we examined whether perinatal exposure to polychlorinated biphenyls (PCBs) and polychlorinated dibenzofurans (PCDFs) induces sustained methylation changes lasting to early adulthood. We used the Illumina HumanMethylation450 BeadChip to assess DNA methylation in whole blood among Yucheng second generation (people perinatal exposed to high PCBs and PCDFs) compared with referents. Thirty male offspring from the Yucheng cohort were randomly selected and matched with 30 male offspring from the Yucheng' neighborhood referents with similar backgrounds. Methylation differences between the Yucheng second generation and non-exposed referents were identified using a P value < 1.06 × 10-7. Differential DNA methylation with epigenome-wide statistical significance was observed for 20 CpGs mapped to 11 genes, and 19 CpGs were correlated with gestational levels of PCBs or PCDF toxic equivalency (PCDF-TEQ) with the same direction of effect. Among the 11 genes, AHRR and CYP1A1 are involved in the aryl hydrocarbon receptor signaling pathway known to mediate dioxin toxicity. MYO1G, FRMD4A, ARL4C, OLFM1, and WWC3 were previously reported to be related to carcinogenesis. This is the first study examining genome-wide DNA methylation among people perinatally exposed to high concentrations of PCBs and PCDFs. We observed novel differential methylation of several genes, indicating that modifications of DNA methylation associated with perinatal PCB and PCDF exposure may persist in exposed offspring for more than 20 years. Furthermore, involvement of several carcinogesis-related genes suggested a potential in utero epigenetic mechanisms.
Collapse
Affiliation(s)
- Kang-Yi Su
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 100, Taiwan; NTU Center for Genomic Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Ming-Chieh Li
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, 350, Taiwan; Department of Public Health, China Medical University College of Public Health, Taichung 404, Taiwan
| | - Nian-Wei Lee
- Institute of Occupational Medicine and Industrial Hygiene, National Taiwan University College of Public Health, Taipei, 100, Taiwan; Department of Environmental and Occupational Medicine, National Taiwan University Hospital, Yun-Lin Branch, Yunlin, 640, Taiwan
| | - Bing-Ching Ho
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 100, Taiwan; NTU Center for Genomic Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Chiou-Ling Cheng
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; NTU Center for Genomic Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Yu-Chen Chuang
- Department of Environmental and Occupational Medicine, National Taiwan University and National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Sung-Liang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 100, Taiwan; NTU Center for Genomic Medicine, National Taiwan University College of Medicine, Taipei 100, Taiwan; Department of Pathology and Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan; Institute of Medical Device and Imaging, College of Medicine, National Taiwan University, Taipei, 100, Taiwan; Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, 100, Taiwan.
| | - Yue Leon Guo
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, 350, Taiwan; Institute of Occupational Medicine and Industrial Hygiene, National Taiwan University College of Public Health, Taipei, 100, Taiwan; Department of Environmental and Occupational Medicine, National Taiwan University and National Taiwan University Hospital, Taipei, 100, Taiwan.
| |
Collapse
|
72
|
Zakarya R, Adcock I, Oliver BG. Epigenetic impacts of maternal tobacco and e-vapour exposure on the offspring lung. Clin Epigenetics 2019; 11:32. [PMID: 30782202 PMCID: PMC6381655 DOI: 10.1186/s13148-019-0631-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/11/2019] [Indexed: 12/15/2022] Open
Abstract
In utero exposure to tobacco products, whether maternal or environmental, have harmful effects on first neonatal and later adult respiratory outcomes. These effects have been shown to persist across subsequent generations, regardless of the offsprings' smoking habits. Established epigenetic modifications induced by in utero exposure are postulated as the mechanism underlying the inherited poor respiratory outcomes. As e-cigarette use is on the rise, their potential to induce similar functional respiratory deficits underpinned by an alteration in the foetal epigenome needs to be explored. This review will focus on the functional and epigenetic impact of in utero exposure to maternal cigarette smoke, maternal environmental tobacco smoke, environmental tobacco smoke and e-cigarette vapour on foetal respiratory outcomes.
Collapse
Affiliation(s)
- Razia Zakarya
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Ian Adcock
- Airway Diseases Section, National Heart and Lung Institute, Imperial College London, London, UK
- Biomedical Research Unit, Section of Respiratory Diseases, Royal Brompton and Harefield NHS Trust, London, UK
| | - Brian G Oliver
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia.
- School of Life Sciences, University of Technology Sydney, Sydney, Australia.
| |
Collapse
|
73
|
Kemp Jacobsen K, Johansen JS, Mellemgaard A, Bojesen SE. AHRR (cg05575921) methylation extent of leukocyte DNA and lung cancer survival. PLoS One 2019; 14:e0211745. [PMID: 30730943 PMCID: PMC6366765 DOI: 10.1371/journal.pone.0211745] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/26/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Prior studies have shown that AHRR (cg05575921) hypomethylation may be a marker of smoking, lung cancer risk and potentially lung cancer survival (in some lung cancer subtypes). It is unknown if AHRR (cg05575921) hypomethylation is associated with reduced survival among lung cancer patients. METHODS In bisulfite treated leukocyte DNA from 465 lung cancer patients from the Copenhagen prospective lung cancer study, we measured AHRR (cg05575921) methylation. 380 died during max follow-up of 4.4 years. Cox proportional hazard models were used to analyze survival as a function of AHRR (cg05575921) methylation. RESULTS We observed the expected inverse correlation between cumulative smoking and AHRR methylation, as methylation (%) decreased (Coefficient -0.03; 95% confidence interval, -0.04- -0.02, p = 8.6x10-15) for every pack-year. Cumulative smoking > 60 pack-years was associated with reduced survival (hazard ratio and 95% confidence interval 1.48; 1.05-2.09), however, AHRR (cg05575921) methylation was not associated with survival when adjusted for sex, body mass index, smoking status, ethnicity, performance status, TNM Classification, and histology type of lung cancer. CONCLUSION AHRR (cg05575921) methylation is linked to smoking but does not provide independent prognostic information in lung cancer patients.
Collapse
Affiliation(s)
- Katja Kemp Jacobsen
- Department of Technology, Faculty of Health and Technology, University College Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Jakob Sidenius Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Anders Mellemgaard
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Stig Egil Bojesen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, and Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
74
|
Zaghlool SB, Mook-Kanamori DO, Kader S, Stephan N, Halama A, Engelke R, Sarwath H, Al-Dous EK, Mohamoud YA, Roemisch-Margl W, Adamski J, Kastenmüller G, Friedrich N, Visconti A, Tsai PC, Spector T, Bell JT, Falchi M, Wahl A, Waldenberger M, Peters A, Gieger C, Pezer M, Lauc G, Graumann J, Malek JA, Suhre K. Deep molecular phenotypes link complex disorders and physiological insult to CpG methylation. Hum Mol Genet 2019; 27:1106-1121. [PMID: 29325019 PMCID: PMC5886112 DOI: 10.1093/hmg/ddy006] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/02/2018] [Indexed: 01/12/2023] Open
Abstract
Epigenetic regulation of cellular function provides a mechanism for rapid organismal adaptation to changes in health, lifestyle and environment. Associations of cytosine-guanine di-nucleotide (CpG) methylation with clinical endpoints that overlap with metabolic phenotypes suggest a regulatory role for these CpG sites in the body's response to disease or environmental stress. We previously identified 20 CpG sites in an epigenome-wide association study (EWAS) with metabolomics that were also associated in recent EWASs with diabetes-, obesity-, and smoking-related endpoints. To elucidate the molecular pathways that connect these potentially regulatory CpG sites to the associated disease or lifestyle factors, we conducted a multi-omics association study including 2474 mass-spectrometry-based metabolites in plasma, urine and saliva, 225 NMR-based lipid and metabolite measures in blood, 1124 blood-circulating proteins using aptamer technology, 113 plasma protein N-glycans and 60 IgG-glyans, using 359 samples from the multi-ethnic Qatar Metabolomics Study on Diabetes (QMDiab). We report 138 multi-omics associations at these CpG sites, including diabetes biomarkers at the diabetes-associated TXNIP locus, and smoking-specific metabolites and proteins at multiple smoking-associated loci, including AHRR. Mendelian randomization suggests a causal effect of metabolite levels on methylation of obesity-associated CpG sites, i.e. of glycerophospholipid PC(O-36: 5), glycine and a very low-density lipoprotein (VLDL-A) on the methylation of the obesity-associated CpG loci DHCR24, MYO5C and CPT1A, respectively. Taken together, our study suggests that multi-omics-associated CpG methylation can provide functional read-outs for the underlying regulatory response mechanisms to disease or environmental insults.
Collapse
Affiliation(s)
- Shaza B Zaghlool
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, PO Box 24144, Doha, Qatar.,Computer Engineering Department, Virginia Tech, Blacksburg, VA 24061, USA
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Centre, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Sara Kader
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, PO Box 24144, Doha, Qatar
| | - Nisha Stephan
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, PO Box 24144, Doha, Qatar
| | - Anna Halama
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, PO Box 24144, Doha, Qatar
| | - Rudolf Engelke
- Proteomics Core, Weill Cornell Medicine-Qatar, Education City, PO Box 24144, Doha, Qatar
| | - Hina Sarwath
- Proteomics Core, Weill Cornell Medicine-Qatar, Education City, PO Box 24144, Doha, Qatar
| | - Eman K Al-Dous
- Genomics Core, Weill Cornell Medicine-Qatar, Education City, PO Box 24144, Doha, Qatar
| | - Yasmin A Mohamoud
- Genomics Core, Weill Cornell Medicine-Qatar, Education City, PO Box 24144, Doha, Qatar
| | - Werner Roemisch-Margl
- Institute of Bioinformatics and Systems Biology, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse, 85764 Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse, 85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstrasse, 85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Alessia Visconti
- Department of Twin Research & Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Pei-Chien Tsai
- Department of Twin Research & Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Tim Spector
- Department of Twin Research & Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Jordana T Bell
- Department of Twin Research & Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Mario Falchi
- Department of Twin Research & Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Annika Wahl
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, D-85764 Neuherberg, Bavaria, Germany.,Institute of Epidemiology II, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, D-85764 Neuherberg, Bavaria, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, D-85764 Neuherberg, Bavaria, Germany.,Institute of Epidemiology II, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, D-85764 Neuherberg, Bavaria, Germany
| | - Annette Peters
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, D-85764 Neuherberg, Bavaria, Germany.,Institute of Epidemiology II, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, D-85764 Neuherberg, Bavaria, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Bavaria, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, D-85764 Neuherberg, Bavaria, Germany.,Institute of Epidemiology II, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, D-85764 Neuherberg, Bavaria, Germany
| | - Marija Pezer
- Glycoscience Research Laboratory, Genos Ltd, HR-10000, Zagreb, Croatia
| | - Gordan Lauc
- Glycoscience Research Laboratory, Genos Ltd, HR-10000, Zagreb, Croatia
| | - Johannes Graumann
- Proteomics Core, Weill Cornell Medicine-Qatar, Education City, PO Box 24144, Doha, Qatar.,Scientific Service Group Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, W.G. Kerckhoff Institute, 61231 Bad Nauheim, Germany
| | - Joel A Malek
- Genomics Core, Weill Cornell Medicine-Qatar, Education City, PO Box 24144, Doha, Qatar
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, PO Box 24144, Doha, Qatar
| |
Collapse
|
75
|
Andersen AM, Lei MK, Philibert RA, Beach SRH. Methylation of MTHFR Moderates the Effect of Smoking on Genomewide Methylation Among Middle Age African Americans. Front Genet 2018; 9:622. [PMID: 30619455 PMCID: PMC6296342 DOI: 10.3389/fgene.2018.00622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/23/2018] [Indexed: 12/24/2022] Open
Abstract
Differential methylation at MTHFR (mMTHFR) has been examined previously as a moderator of changes in methylation among nascent smokers, but the effects of mMTHFR on genomewide patterns of methylation among established smokers in middle age are unknown. In the current investigation we examined a sample of 180 African American middle-aged smokers and non-smokers to test for patterns indicative of three different potential mechanisms of impact on epigenetic remodeling in response to long-term smoking. We found that mMTHFR moderated the association between smoking and changes in methylation for more than 25% of the 909 loci previously identified as being associated with smoking at a genomewide level of significance in middle-aged African Americans. Observed patterns of effect indicated amplification of both hyper and hypo methylating responses to smoking among those with lower mMTHFR. Moderating effects were robust to controls for sex, age, diet, and cell-type variation. Implications for potential mechanisms conferring effects are discussed. Of particular potential practical importance was a strong effect of mMTHFR on hypomethylation at GPR15 in response to smoking, indicative of the differential impact of MTHFR activity on changes in a specific cell population linked to inflammatory disease in smokers.
Collapse
Affiliation(s)
- Allan M Andersen
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States
| | - Man-Kit Lei
- Department of Sociology, University of Georgia, Athens, GA, United States.,Center for Family Research, University of Georgia, Athens, GA, United States
| | - Robert A Philibert
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States.,Behavioral Diagnostics, Coralville, IA, United States
| | - Steven R H Beach
- Center for Family Research, University of Georgia, Athens, GA, United States.,Department of Psychology, University of Georgia, Athens, GA, United States
| |
Collapse
|
76
|
Tsai PC, Glastonbury CA, Eliot MN, Bollepalli S, Yet I, Castillo-Fernandez JE, Carnero-Montoro E, Hardiman T, Martin TC, Vickers A, Mangino M, Ward K, Pietiläinen KH, Deloukas P, Spector TD, Viñuela A, Loucks EB, Ollikainen M, Kelsey KT, Small KS, Bell JT. Smoking induces coordinated DNA methylation and gene expression changes in adipose tissue with consequences for metabolic health. Clin Epigenetics 2018; 10:126. [PMID: 30342560 PMCID: PMC6196025 DOI: 10.1186/s13148-018-0558-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 10/03/2018] [Indexed: 12/14/2022] Open
Abstract
Background Tobacco smoking is a risk factor for multiple diseases, including cardiovascular disease and diabetes. Many smoking-associated signals have been detected in the blood methylome, but the extent to which these changes are widespread to metabolically relevant tissues, and impact gene expression or metabolic health, remains unclear. Methods We investigated smoking-associated DNA methylation and gene expression variation in adipose tissue biopsies from 542 healthy female twins. Replication, tissue specificity, and longitudinal stability of the smoking-associated effects were explored in additional adipose, blood, skin, and lung samples. We characterized the impact of adipose tissue smoking methylation and expression signals on metabolic disease risk phenotypes, including visceral fat. Results We identified 42 smoking-methylation and 42 smoking-expression signals, where five genes (AHRR, CYP1A1, CYP1B1, CYTL1, F2RL3) were both hypo-methylated and upregulated in current smokers. CYP1A1 gene expression achieved 95% prediction performance of current smoking status. We validated and replicated a proportion of the signals in additional primary tissue samples, identifying tissue-shared effects. Smoking leaves systemic imprints on DNA methylation after smoking cessation, with stronger but shorter-lived effects on gene expression. Metabolic disease risk traits such as visceral fat and android-to-gynoid ratio showed association with methylation at smoking markers with functional impacts on expression, such as CYP1A1, and at tissue-shared smoking signals, such as NOTCH1. At smoking-signals, BHLHE40 and AHRR DNA methylation and gene expression levels in current smokers were predictive of future gain in visceral fat upon smoking cessation. Conclusions Our results provide the first comprehensive characterization of coordinated DNA methylation and gene expression markers of smoking in adipose tissue. The findings relate to human metabolic health and give insights into understanding the widespread health consequence of smoking outside of the lung. Electronic supplementary material The online version of this article (10.1186/s13148-018-0558-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pei-Chien Tsai
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK. .,Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan. .,Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan.
| | - Craig A Glastonbury
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK.,Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, OX3 7LF, UK
| | - Melissa N Eliot
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, 02912, USA
| | - Sailalitha Bollepalli
- Institute for Molecular Medicine Finland (FIMM) and Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Idil Yet
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK.,Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, 06100, Ankara, Turkey
| | | | - Elena Carnero-Montoro
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK.,Pfizer - University of Granada - Andalusian Government Center for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Thomas Hardiman
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK.,Division of Cancer Studies, King's College London, London, SE1 9RT, UK
| | - Tiphaine C Martin
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, 10029, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, 10029, USA
| | - Alice Vickers
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK.,Centre for Stem Cells and Regenerative Medicine, King's College London, Floor 28, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK.,NIHR Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, SE1 9RT, UK
| | - Kirsten Ward
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK
| | - Kirsi H Pietiläinen
- Research Programs Unit, Diabetes and Obesity, Obesity Research Unit, University of Helsinki, Helsinki, Finland.,Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK.,Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK
| | - Ana Viñuela
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK.,Department of Genetic Medicine and Development, University of Geneva Medical School, 1211, Geneva, Switzerland.,Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, 1211, Geneva, Switzerland.,Swiss Institute of Bioinformatics, 1211, Geneva, Switzerland
| | - Eric B Loucks
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, 02912, USA
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland (FIMM) and Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Karl T Kelsey
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, 02912, USA.,Department of Laboratory Medicine & Pathology, Brown University, Providence, RI, 02912, USA
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK
| | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK.
| |
Collapse
|
77
|
Aggarwal T, Wadhwa R, Thapliyal N, Sharma K, Rani V, Maurya PK. Oxidative, inflammatory, genetic, and epigenetic biomarkers associated with chronic obstructive pulmonary disorder. J Cell Physiol 2018; 234:2067-2082. [DOI: 10.1002/jcp.27181] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/17/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Taru Aggarwal
- Amity Institute of Biotechnology, Amity UniversityNoida India
| | - Ridhima Wadhwa
- Amity Institute of Biotechnology, Amity UniversityNoida India
| | | | - Kanishka Sharma
- Amity Education GroupOakdale, Long Island (Suffolk) New York
| | - Varsha Rani
- Amity Education GroupOakdale, Long Island (Suffolk) New York
| | - Pawan K. Maurya
- Amity Institute of Biotechnology, Amity UniversityNoida India
- Amity Education GroupOakdale, Long Island (Suffolk) New York
- Interdisciplinary Laboratory of Clinical Neuroscience (LINC), Department of PsychiatryFederal University of São PauloSão Paulo Brazil
| |
Collapse
|
78
|
Smoking is Associated to DNA Methylation in Atherosclerotic Carotid Lesions. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2018; 11:e002030. [DOI: 10.1161/circgen.117.002030] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
79
|
Park SL, Patel YM, Loo LWM, Mullen DJ, Offringa IA, Maunakea A, Stram DO, Siegmund K, Murphy SE, Tiirikainen M, Le Marchand L. Association of internal smoking dose with blood DNA methylation in three racial/ethnic populations. Clin Epigenetics 2018; 10:110. [PMID: 30139389 PMCID: PMC6108111 DOI: 10.1186/s13148-018-0543-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/06/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related death. While cigarette smoking is the primary cause of this malignancy, risk differs across racial/ethnic groups. For the same number of cigarettes smoked, Native Hawaiians compared to whites are at greater risk and Japanese Americans are at lower risk of developing lung cancer. DNA methylation of specific CpG sites (e.g., in AHRR and F2RL3) is the most common blood epigenetic modification associated with smoking status. However, the influence of internal smoking dose, measured by urinary nicotine equivalents (NE), on DNA methylation in current smokers has not been investigated, nor has a study evaluated whether for the same smoking dose, circulating leukocyte DNA methylation patterns differ by race. METHODS We conducted an epigenome-wide association study (EWAS) of NE in 612 smokers from three racial/ethnic groups: whites (n = 204), Native Hawaiians (n = 205), and Japanese Americans (n = 203). Genome-wide DNA methylation profiling of blood leukocyte DNA was measured using the Illumina 450K BeadChip array. Average β value, the ratio of signal from a methylated probe relative to the sum of the methylated and unmethylated probes at that CpG, was the dependent variables in linear regression models adjusting for age, sex, race (for pan-ethnic analysis), and estimated cell-type distribution. RESULTS We found that NE was significantly associated with six differentially methylated CpG sites (Bonferroni corrected p < 1.48 × 10-7): four in or near the FOXK2, PBX1, FNDC7, and FUBP3 genes and two in non-annotated genetic regions. Higher levels of NE were associated with increasing methylation beta-valuesin all six sites. For all six CpG sites, the association was only observed in Native Hawaiians, suggesting that the influence of smoking dose on DNA methylation patterns is heterogeneous across race/ethnicity (p interactions < 8.8 × 10-8). We found two additional CpG sites associated with NE in only Native Hawaiians. CONCLUSIONS In conclusion, internal smoking dose was associated with increased DNA methylation in circulating leukocytes at specific sites in Native Hawaiian smokers but not in white or Japanese American smokers.
Collapse
Affiliation(s)
- Sungshim L Park
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1450 Biggy Street, NRT 1509G, Los Angeles, CA, 90033, USA.
| | - Yesha M Patel
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1450 Biggy Street, NRT 1509G, Los Angeles, CA, 90033, USA
| | - Lenora W M Loo
- Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Daniel J Mullen
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90032, USA
| | - Ite A Offringa
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90032, USA
| | - Alika Maunakea
- University of Hawaii John A. Burns School of Medicine, Honolulu, HI, 96813, USA
| | - Daniel O Stram
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1450 Biggy Street, NRT 1509G, Los Angeles, CA, 90033, USA
| | - Kimberly Siegmund
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1450 Biggy Street, NRT 1509G, Los Angeles, CA, 90033, USA
| | - Sharon E Murphy
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Maarit Tiirikainen
- Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, USA.
| |
Collapse
|
80
|
Murphy SE, Park SL, Balbo S, Haiman CA, Hatsukami DK, Patel Y, Peterson LA, Stepanov I, Stram DO, Tretyakova N, Hecht SS, Le Marchand L. Tobacco biomarkers and genetic/epigenetic analysis to investigate ethnic/racial differences in lung cancer risk among smokers. NPJ Precis Oncol 2018; 2:17. [PMID: 30155522 PMCID: PMC6105591 DOI: 10.1038/s41698-018-0057-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/08/2018] [Accepted: 06/13/2018] [Indexed: 12/31/2022] Open
Abstract
The Multiethnic Cohort Study has demonstrated that African Americans and Native Hawaiians have a higher risk for lung cancer due to cigarette smoking than Whites while Latinos and Japanese Americans have a lower risk. These findings are consistent with other epidemiologic studies in the literature. In this review, we summarize tobacco carcinogen and toxicant biomarker studies and genetic analyses which partially explain these differences. As determined by measurement of total nicotine equivalents in urine, which account for about 85% of the nicotine dose, African Americans take up greater amounts of nicotine than Whites per cigarette while Japanese Americans take up less. There are corresponding differences in the uptake of tobacco smoke carcinogens such as tobacco-specific nitrosamines, polycyclic aromatic hydrocarbons, 1,3-butadiene, and other toxic volatiles. The lower nicotine uptake of Japanese Americans is clearly linked to the preponderance of low activity forms of the primary nicotine metabolizing enzyme CYP2A6 in this ethnic group, leading to more unchanged nicotine in the body and thus lower smoking intensity. But the relatively high risk of Native Hawaiians and the low risk of Latino smokers for lung cancer are not explained by these factors. The possible role of epigenetics in modifying lung cancer risk among smokers is also discussed here. The results of these published studies may lead to a better understanding of susceptibility factors for lung cancer in cigarette smokers thus potentially identifying biomarkers that can detect those individuals at highest risk so that preventive approaches can be initiated at an early stage of the lung cancer development process.
Collapse
Affiliation(s)
- Sharon E. Murphy
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455 USA
| | - Sungshim Lani Park
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089 USA
| | - Silvia Balbo
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455 USA
| | - Christopher A. Haiman
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089 USA
| | | | - Yesha Patel
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089 USA
| | - Lisa A. Peterson
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455 USA
| | - Irina Stepanov
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455 USA
| | - Daniel O. Stram
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089 USA
| | - Natalia Tretyakova
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455 USA
| | - Stephen S. Hecht
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455 USA
| | - Loïc Le Marchand
- Cancer Research Center of Hawaii, University of Hawaii, Honolulu, HI 96813 USA
| |
Collapse
|
81
|
Haase T, Müller C, Krause J, Röthemeier C, Stenzig J, Kunze S, Waldenberger M, Münzel T, Pfeiffer N, Wild PS, Michal M, Marini F, Karakas M, Lackner KJ, Blankenberg S, Zeller T. Novel DNA Methylation Sites Influence GPR15 Expression in Relation to Smoking. Biomolecules 2018; 8:biom8030074. [PMID: 30127295 PMCID: PMC6163736 DOI: 10.3390/biom8030074] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/06/2018] [Accepted: 08/09/2018] [Indexed: 11/24/2022] Open
Abstract
Smoking is a major risk factor for cardiovascular diseases and has been implicated in the regulation of the G protein-coupled receptor 15 (GPR15) by affecting CpG methylation. The G protein-coupled receptor 15 is involved in angiogenesis and inflammation. An effect on GPR15 gene regulation has been shown for the CpG site CpG3.98251294. We aimed to analyze the effect of smoking on GPR15 expression and methylation sites spanning the GPR15 locus. DNA methylation of nine GPR15 CpG sites was measured in leukocytes from 1291 population-based individuals using the EpiTYPER. Monocytic GPR15 expression was measured by qPCR at baseline and five-years follow up. GPR15 gene expression was upregulated in smokers (beta (ß) = −2.699, p-value (p) = 1.02 × 10−77) and strongly correlated with smoking exposure (ß = −0.063, p = 2.95 × 10−34). Smoking cessation within five years reduced GPR15 expression about 19% (p = 9.65 × 10−5) with decreasing GPR15 expression over time (ß = 0.031, p = 3.81 × 10−6). Additionally, three novel CpG sites within GPR15 affected by smoking were identified. For CpG3.98251047, DNA methylation increased steadily after smoking cessation (ß = 0.123, p = 1.67 × 10−3) and strongly correlated with changes in GPR15 expression (ß = 0.036, p = 4.86 × 10−5). Three novel GPR15 CpG sites were identified in relation to smoking and GPR15 expression. Our results provide novel insights in the regulation of GPR15, which possibly linked smoking to inflammation and disease progression.
Collapse
Affiliation(s)
- Tina Haase
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
| | - Christian Müller
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
| | - Julia Krause
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
| | - Caroline Röthemeier
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
| | - Justus Stenzig
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany.
| | - Sonja Kunze
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany.
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany.
| | - Melanie Waldenberger
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany.
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany.
| | - Thomas Münzel
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
- Center for Cardiology, Cardiology I, University Medical Center Mainz, Johannes Gutenberg University-Mainz, 55131 Mainz, Germany.
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
- Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Philipp S Wild
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
- Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Matthias Michal
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Federico Marini
- University Medical Center, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), 55131 Mainz, Germany.
| | - Mahir Karakas
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
| | - Karl J Lackner
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany.
| | - Stefan Blankenberg
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
| | - Tanja Zeller
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), 13316 Berlin, Germany.
| |
Collapse
|
82
|
Alghanim H, Wu W, McCord B. DNA methylation assay based on pyrosequencing for determination of smoking status. Electrophoresis 2018; 39:2806-2814. [PMID: 29956353 DOI: 10.1002/elps.201800098] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/30/2018] [Accepted: 06/01/2018] [Indexed: 11/06/2022]
Abstract
The goal of this study was to utilize pyrosequencing to identify CpG sites indicative of tobacco smoking using DNA sequences surrounding ten frequently reported smoking-related CpGs. Initially, six genetic loci were investigated including AHRR, 2q37, 6p21.33, GFI1, F2RL3, and MYO1G in order to detect novel CpG sites associated with tobacco smoking. The methylation data revealed a set of 23 consecutive CpG sites in blood (Chr5:373,115-Chr5:373,653) that were significantly hypomethylated in current smokers. In addition, 10 of these 23 CpGs were also significantly hypomethylated in the saliva of current smokers. The most significant CpG sites were located at Chr5:373,490 in blood and Chr5:373,476 in saliva with a decrease in methylation in current smokers of 42.3% and 21.3% respectively. In the model-building steps of this study, a quick 4-CpG assay was developed. The assay consisted of the top ranked CpG sites in blood and saliva. The assay was applied in a leave-one-out approach to test its ability to infer an individual's self-identified history of smoking habits. A multinomial logistic regression model (MLR) containing all 4 CpG sites gave the most accurate results in blood and saliva. In blood, the model correctly predicted 90.0% of current smokers, 66.7% of former smokers, and 84.9% of never smokers. In addition, the MLR model correctly predicted 86.9% of current smokers, 54.5% of former smokers, and 77.8% of never smokers in saliva. These results demonstrate that this pyrosequencing-based assay can provide an effective tool for identifying individuals who smoke tobacco, particularly when using epigenetic markers in blood.
Collapse
Affiliation(s)
- Hussain Alghanim
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, USA.,General Department of Forensic Science and Criminology, Dubai Police, Dubai, UAE
| | - Wensong Wu
- Department of Mathematics and Statistics, Florida International University, Miami, FL, USA
| | - Bruce McCord
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, USA
| |
Collapse
|
83
|
Jessen WJ, Borgerding MF, Prasad GL. Global methylation profiles in buccal cells of long-term smokers and moist snuff consumers. Biomarkers 2018; 23:625-639. [DOI: 10.1080/1354750x.2018.1466367] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Walter J. Jessen
- Laboratory Corporation of America Holdings (LabCorp), Burlington, NC, USA
| | | | | |
Collapse
|
84
|
Philibert R, Dogan M, Noel A, Miller S, Krukow B, Papworth E, Cowley J, Knudsen A, Beach SR, Black D. Genome-wide and digital polymerase chain reaction epigenetic assessments of alcohol consumption. Am J Med Genet B Neuropsychiatr Genet 2018; 177:479-488. [PMID: 29704316 PMCID: PMC9518929 DOI: 10.1002/ajmg.b.32636] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/13/2018] [Accepted: 03/27/2018] [Indexed: 01/19/2023]
Abstract
The lack of readily employable biomarkers of alcohol consumption is a problem for clinicians and researchers. In 2014, we published a preliminary DNA methylation signature of heavy alcohol consumption that remits as a function of abstinence. Herein, we present new genome-wide methylation findings from a cohort of additional subjects and a meta-analysis of the data. Using DNA from 47 consecutive heavy drinkers admitted for alcohol detoxification in the context of alcohol treatment and 47 abstinent controls, we replicate the 2014 results and show that 21,221 CpG residues are differentially methylated in active heavy drinkers. Meta-analysis of all data from the 448,058 probes common to the two methylation platforms shows a similarly profound signature with confirmation of findings from other groups. Principal components analyses show that genome-wide methylation changes in response to alcohol consumption load on two major factors with one component accounting at least 50% of the total variance in both smokers and nonsmoking alcoholics. Using data from the arrays, we derive a panel of five methylation probes that classifies use status with a receiver operator characteristic area under the curve (AUC) of 0.97. Finally, using droplet digital polymerase chain reaction (PCR), we convert these array-based findings to two marker assays with an AUC of 0.95 and a four marker set AUC of 0.98. We conclude that DNA methylation assessments are capable of quantifying alcohol use status and suggest that readily employable digital PCR approaches for substance consumption may find widespread use in alcohol-related research and patient care.
Collapse
Affiliation(s)
- Robert Philibert
- Behavioral Diagnostics, Coralville, Iowa 52241
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242
| | - Meesha Dogan
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242
- Cardio Diagnostics, Coralville, Iowa 52241
| | - Amanda Noel
- Behavioral Diagnostics, Coralville, Iowa 52241
| | | | | | | | - Joseph Cowley
- Center for Alcohol and Drug Services, Davenport, IA 52804
| | | | | | - Donald Black
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
85
|
Wan M, Bennett BD, Pittman GS, Campbell MR, Reynolds LM, Porter DK, Crowl CL, Wang X, Su D, Englert NA, Thompson IJ, Liu Y, Bell DA. Identification of Smoking-Associated Differentially Methylated Regions Using Reduced Representation Bisulfite Sequencing and Cell type-Specific Enhancer Activation and Gene Expression. ENVIRONMENTAL HEALTH PERSPECTIVES 2018; 126:047015. [PMID: 29706059 PMCID: PMC6071796 DOI: 10.1289/ehp2395] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 03/22/2018] [Accepted: 03/22/2018] [Indexed: 05/23/2023]
Abstract
BACKGROUND Cigarette smoke is a causal factor in cancers and cardiovascular disease. Smoking-associated differentially methylated regions (SM-DMRs) have been observed in disease studies, but the causal link between altered DNA methylation and transcriptional change is obscure. OBJECTIVE Our objectives were to finely resolve SM-DMRs and to interrogate the mechanistic link between SM-DMRs and altered transcription of enhancer noncoding RNA (eRNA) and mRNA in human circulating monocytes. METHOD We integrated SM-DMRs identified by reduced representation bisulfite sequencing (RRBS) of circulating CD14+ monocyte DNA collected from two independent human studies [n=38 from Clinical Research Unit (CRU) and n=55 from the Multi-Ethnic Study of Atherosclerosis (MESA), about half of whom were active smokers] with gene expression for protein-coding genes and noncoding RNAs measured by RT-PCR or RNA sequencing. Candidate SM-DMRs were compared with RRBS of purified CD4+ T cells, CD8+ T cells, CD15+ granulocytes, CD19+ B cells, and CD56+ NK cells (n=19 females, CRU). DMRs were validated using pyrosequencing or bisulfite amplicon sequencing in up to 85 CRU volunteers, who also provided saliva DNA. RESULTS RRBS identified monocyte SM-DMRs frequently located in putative gene regulatory regions. The most significant monocyte DMR occurred at a poised enhancer in the aryl-hydrocarbon receptor repressor gene (AHRR) and it was also detected in both granulocytes and saliva DNA. To our knowledge, we identify for the first time that SM-DMRs in or near AHRR, C5orf55-EXOC-AS, and SASH1 were associated with increased noncoding eRNA as well as mRNA in monocytes. Functionally, the AHRR SM-DMR appeared to up-regulate AHRR mRNA through activating the AHRR enhancer, as suggested by increased eRNA in the monocytes, but not granulocytes, from smokers compared with nonsmokers. CONCLUSIONS Our findings suggest that AHRR SM-DMR up-regulates AHRR mRNA in a monocyte-specific manner by activating the AHRR enhancer. Cell type-specific activation of enhancers at SM-DMRs may represent a mechanism driving smoking-related disease. https://doi.org/10.1289/EHP2395.
Collapse
Affiliation(s)
- Ma Wan
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Brian D Bennett
- Integrative Bioinformatics Support Group, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Gary S Pittman
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Michelle R Campbell
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Lindsay M Reynolds
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Devin K Porter
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Christopher L Crowl
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Xuting Wang
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Dan Su
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Neal A Englert
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Isabel J Thompson
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Douglas A Bell
- Environmental Epigenomics and Disease Group, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| |
Collapse
|
86
|
Philibert R, Dogan M, Noel A, Miller S, Krukow B, Papworth E, Cowley J, Long JD, Beach SRH, Black DW. Dose Response and Prediction Characteristics of a Methylation Sensitive Digital PCR Assay for Cigarette Consumption in Adults. Front Genet 2018; 9:137. [PMID: 29740475 PMCID: PMC5928210 DOI: 10.3389/fgene.2018.00137] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/03/2018] [Indexed: 01/16/2023] Open
Abstract
The tobacco use disorders are the largest preventable cause of morbidity and mortality in the world. A substantial barrier to the development of better intervention and screening measures is the lack of clinically employable biomarkers to detect the existence and extent of tobacco consumption. In prior work, we and others have shown that array based assessment of DNA methylation status at cg05575921 is a sensitive and quantitative method for assessing cigarette consumption. Unfortunately, in general, arrays are not practical clinical tools. Herein, we detail the prediction performance metrics and dose dependency of a clinically implementable droplet digital PCR (ddPCR) assay for cigarette consumption in adults. First, we demonstrate that measurements of cg05575921 as determined by Illumina array and ddPCR are highly correlated (R2 = 0.98, n = 92). Second, using clinical data and biomaterial from 177 subjects ranging from 18 to 78 years of age, we show that the Receiver Operating Characteristic (ROC) area under the curve (AUC) for classifying smoking status using methylation status at cg05575921 is 0.99. Finally, we conduct modeling analyses of cigarette consumption over discrete time periods to show that methylation status is best correlated with mean cigarette consumption over the past year (R2 = 0.5) and that demethylation at cg05575921 is dose dependent with a demethylation (delta beta) of 1% being equivalent to 1.2 cigarettes per day. But we do not find a relationship between Fagerstrom score and DNA methylation. We conclude that ddPCR assessment of cg05575921 methylation is an accurate method for assessing the presence and extent of cigarette consumption in adult subjects. We suggest that skillful clinical implementation of this approach alone or in combination with other assessment methods could lead to substantial reduction of cigarette consumption related morbidity and mortality.
Collapse
Affiliation(s)
- Robert Philibert
- Behavioral Diagnostics LLC, Coralville, IA, United States
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States
| | - Meesha Dogan
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States
- Cardio Diagnostics, Coralville, IA, United States
| | - Amanda Noel
- Behavioral Diagnostics LLC, Coralville, IA, United States
| | - Shelly Miller
- Behavioral Diagnostics LLC, Coralville, IA, United States
| | - Brianna Krukow
- Behavioral Diagnostics LLC, Coralville, IA, United States
| | - Emma Papworth
- Behavioral Diagnostics LLC, Coralville, IA, United States
| | - Joseph Cowley
- Center for Alcohol and Drug Services, Davenport, IA, United States
| | | | - Steven R. H. Beach
- Center for Family Studies at the University of Georgia, Athens, GA, United States
| | - Donald W. Black
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
87
|
Kopa PN, Pawliczak R. Effect of smoking on gene expression profile – overall mechanism, impact on respiratory system function, and reference to electronic cigarettes. Toxicol Mech Methods 2018; 28:397-409. [DOI: 10.1080/15376516.2018.1461289] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Paulina Natalia Kopa
- Department of Immunopathology, Division of Allergology, Immunology and Dermatology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz, Lodz, Poland
| | - Rafał Pawliczak
- Department of Immunopathology, Division of Allergology, Immunology and Dermatology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
88
|
Suitability of biomarkers of biological effects (BOBEs) for assessing the likelihood of reducing the tobacco related disease risk by new and innovative tobacco products: A literature review. Regul Toxicol Pharmacol 2018; 94:203-233. [DOI: 10.1016/j.yrtph.2018.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 02/04/2018] [Accepted: 02/05/2018] [Indexed: 02/07/2023]
|
89
|
Kodal JB, Kobylecki CJ, Vedel-Krogh S, Nordestgaard BG, Bojesen SE. AHRR hypomethylation, lung function, lung function decline and respiratory symptoms. Eur Respir J 2018; 51:13993003.01512-2017. [PMID: 29348151 DOI: 10.1183/13993003.01512-2017] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 12/31/2017] [Indexed: 12/22/2022]
Abstract
Epigenome-wide association studies have shown a consistent association between smoking exposure and hypomethylation in the aryl hydrocarbon receptor repressor (AHRR) gene (cg05575921). We tested the hypothesis that AHRR hypomethylation is associated with low lung function, steeper lung function decline, and respiratory symptoms in the general population.AHRR methylation extent was measured in 9113 individuals from the 1991-1994 examination of the Copenhagen City Heart Study, using bisulfite-treated leukocyte DNA. Spirometry at the time of blood sampling was available for all individuals. Lung function was measured again for 4532 of these individuals in 2001-2003.Cross-sectionally, a 10% lower methylation extent was associated with a 0.2 z-score (95% CI 0.1-0.2) lower forced expiratory volume in 1 s (FEV1) after multivariable adjustment including smoking. Hypomethylation was also associated with a lower z-score for both forced vital capacity (FVC) and FEV1/FVC. In prospective analyses, individuals in the lowest versus highest tertile of methylation extent had a steeper decline in FEV1/height3 (p for examination×methylation interaction=0.003) and FVC/height3 (p=0.01), but not FEV1/FVC (p=0.08). Multivariable-adjusted odds ratios per 10% lower methylation extent were 1.31 (95% CI 1.18-1.45) for chronic bronchitis and 1.21 (95% CI 1.13-1.30) for any respiratory symptoms.AHRR hypomethylation was associated with low lung function, steeper lung function decline, and respiratory symptoms.
Collapse
Affiliation(s)
- Jakob B Kodal
- Dept of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Camilla J Kobylecki
- Dept of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Signe Vedel-Krogh
- Dept of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Dept of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Stig E Bojesen
- Dept of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark .,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
90
|
Wahl A, Kasela S, Carnero-Montoro E, van Iterson M, Štambuk J, Sharma S, van den Akker E, Klaric L, Benedetti E, Razdorov G, Trbojević-Akmačić I, Vučković F, Ugrina I, Beekman M, Deelen J, van Heemst D, Heijmans BT, B.I.O.S. Consortium, Wuhrer M, Plomp R, Keser T, Šimurina M, Pavić T, Gudelj I, Krištić J, Grallert H, Kunze S, Peters A, Bell JT, Spector TD, Milani L, Slagboom PE, Lauc G, Gieger C. IgG glycosylation and DNA methylation are interconnected with smoking. Biochim Biophys Acta Gen Subj 2018; 1862:637-648. [DOI: 10.1016/j.bbagen.2017.10.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/01/2017] [Accepted: 10/16/2017] [Indexed: 01/18/2023]
|
91
|
Knopik VS, Marceau K, Bidwell LC, Rolan E. Prenatal substance exposure and offspring development: Does DNA methylation play a role? Neurotoxicol Teratol 2018; 71:50-63. [PMID: 29408446 DOI: 10.1016/j.ntt.2018.01.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 01/12/2018] [Accepted: 01/24/2018] [Indexed: 12/17/2022]
Abstract
The period of in utero development is one of the most critical windows during which adverse conditions and exposures may influence the growth and development of the fetus as well as its future postnatal health and behavior. Maternal substance use during pregnancy remains a relatively common but nonetheless hazardous in utero exposure. For example, previous epidemiological studies have associated prenatal substance exposure with reduced birth weight, poor developmental and psychological outcomes, and increased risk for diseases and behavioral disorders (e.g., externalizing behaviors like ADHD, conduct disorder, and substance use) later in life. Researchers are now learning that many of the mechanisms whereby adverse in utero exposures may affect key pathways crucial for proper fetal growth and development are epigenetic in nature, with the majority of work in humans considering DNA methylation specifically. This review will explore the research to date on epigenetic alterations tied to maternal substance use during pregnancy and will also discuss the possible role of DNA methylation in the robust relationship between maternal substance use and later behavioral and developmental sequelae in offspring.
Collapse
Affiliation(s)
- Valerie S Knopik
- Department of Human Development and Family Studies, Purdue University, West Lafayette, IN, USA.
| | - Kristine Marceau
- Department of Human Development and Family Studies, Purdue University, West Lafayette, IN, USA
| | - L Cinnamon Bidwell
- Institute of Cognitive Science, University of Colorado, Boulder, CO, USA
| | - Emily Rolan
- Department of Human Development and Family Studies, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
92
|
Wu DD, Song J, Bartel S, Krauss-Etschmann S, Rots MG, Hylkema MN. The potential for targeted rewriting of epigenetic marks in COPD as a new therapeutic approach. Pharmacol Ther 2018; 182:1-14. [PMID: 28830839 DOI: 10.1016/j.pharmthera.2017.08.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is an age and smoking related progressive, pulmonary disorder presenting with poorly reversible airflow limitation as a result of chronic bronchitis and emphysema. The prevalence, disease burden for the individual, and mortality of COPD continues to increase, whereas no effective treatment strategies are available. For many years now, a combination of bronchodilators and anti-inflammatory corticosteroids has been most widely used for therapeutic management of patients with persistent COPD. However, this approach has had disappointing results as a large number of COPD patients are corticosteroid resistant. In patients with COPD, there is emerging evidence showing aberrant expression of epigenetic marks such as DNA methylation, histone modifications and microRNAs in blood, sputum and lung tissue. Therefore, novel therapeutic approaches may exist using epigenetic therapy. This review aims to describe and summarize current knowledge of aberrant expression of epigenetic marks in COPD. In addition, tools available for restoration of epigenetic marks are described, as well as delivery mechanisms of epigenetic editors to cells. Targeting epigenetic marks might be a very promising tool for treatment and lung regeneration in COPD in the future.
Collapse
Affiliation(s)
- Dan-Dan Wu
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands; University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, P.R. China
| | - Juan Song
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands; University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands; Tianjin Medical University, School of Basic Medical Sciences, Department of Biochemistry and Molecular Biology, Department of Immunology, Tianjin, China
| | - Sabine Bartel
- Early Life Origins of Chronic Lung Disease, Priority Area Asthma & Allergy, Leibnitz Center for Medicine and Biosciences, Research Center Borstel and Christian Albrechts University Kiel; Airway Research Center North, member of the German Center for Lung Research (DZL), Germany
| | - Susanne Krauss-Etschmann
- Early Life Origins of Chronic Lung Disease, Priority Area Asthma & Allergy, Leibnitz Center for Medicine and Biosciences, Research Center Borstel and Christian Albrechts University Kiel; Airway Research Center North, member of the German Center for Lung Research (DZL), Germany
| | - Marianne G Rots
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Machteld N Hylkema
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands; University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands.
| |
Collapse
|
93
|
Crime investigation through DNA methylation analysis: methods and applications in forensics. EGYPTIAN JOURNAL OF FORENSIC SCIENCES 2018. [DOI: 10.1186/s41935-018-0042-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
94
|
Reynolds LM, Lohman K, Pittman GS, Barr RG, Chi GC, Kaufman J, Wan M, Bell DA, Blaha MJ, Rodriguez CJ, Liu Y. Tobacco exposure-related alterations in DNA methylation and gene expression in human monocytes: the Multi-Ethnic Study of Atherosclerosis (MESA). Epigenetics 2018; 12:1092-1100. [PMID: 29166816 PMCID: PMC5810757 DOI: 10.1080/15592294.2017.1403692] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Alterations in DNA methylation and gene expression in blood leukocytes are potential biomarkers of harm and mediators of the deleterious effects of tobacco exposure. However, methodological issues, including the use of self-reported smoking status and mixed cell types have made previously identified alterations in DNA methylation and gene expression difficult to interpret. In this study, we examined associations of tobacco exposure with DNA methylation and gene expression, utilizing a biomarker of tobacco exposure (urine cotinine) and CD14+ purified monocyte samples from 934 participants of the community-based Multi-Ethnic Study of Atherosclerosis (MESA). Urine cotinine levels were measured using an immunoassay. DNA methylation and gene expression were measured with microarrays. Multivariate linear regression was used to test for associations adjusting for age, sex, race/ethnicity, education, and study site. Urine cotinine levels were associated with methylation of 176 CpGs [false discovery rate (FDR)<0.01]. Four CpGs not previously identified by studies of non-purified blood samples nominally replicated (P value<0.05) with plasma cotinine-associated methylation in 128 independent monocyte samples. Urine cotinine levels associated with expression of 12 genes (FDR<0.01), including increased expression of P2RY6 (Beta ± standard error = 0.078 ± 0.008, P = 1.99 × 10−22), a gene previously identified to be involved in the release of pro-inflammatory cytokines. No cotinine-associated (FDR<0.01) methylation profiles significantly (FDR<0.01) correlated with cotinine-associated (FDR<0.01) gene expression profiles. In conclusion, our findings i) identify potential monocyte-specific smoking-associated methylation patterns and ii) suggest that alterations in methylation may not be a main mechanism regulating gene expression in monocytes in response to cigarette smoking.
Collapse
Affiliation(s)
- Lindsay M Reynolds
- a Division of Public Health Sciences , Wake Forest School of Medicine , Winston-Salem , NC 27157
| | - Kurt Lohman
- a Division of Public Health Sciences , Wake Forest School of Medicine , Winston-Salem , NC 27157
| | - Gary S Pittman
- b Immunity, Inflammation and Disease Laboratory , National Institute of Environmental Health Sciences, National Institutes of Health , Research Triangle Park , NC 27709
| | - R Graham Barr
- c Departments of Medicine and Epidemiology , Columbia University Medical Center , New York , NY 10032
| | - Gloria C Chi
- d Department of Epidemiology, School of Public Health , University of Washington , Seattle , WA 98105
| | - Joel Kaufman
- d Department of Epidemiology, School of Public Health , University of Washington , Seattle , WA 98105
| | - Ma Wan
- b Immunity, Inflammation and Disease Laboratory , National Institute of Environmental Health Sciences, National Institutes of Health , Research Triangle Park , NC 27709
| | - Douglas A Bell
- b Immunity, Inflammation and Disease Laboratory , National Institute of Environmental Health Sciences, National Institutes of Health , Research Triangle Park , NC 27709
| | - Michael J Blaha
- e Johns Hopkins Ciccarone Center for the Prevention of Heart Disease , Baltimore , MD 21287
| | - Carlos J Rodriguez
- a Division of Public Health Sciences , Wake Forest School of Medicine , Winston-Salem , NC 27157
| | - Yongmei Liu
- a Division of Public Health Sciences , Wake Forest School of Medicine , Winston-Salem , NC 27157
| |
Collapse
|
95
|
Gao X, Zhang Y, Breitling LP, Brenner H. Relationship of tobacco smoking and smoking-related DNA methylation with epigenetic age acceleration. Oncotarget 2018; 7:46878-46889. [PMID: 27276709 PMCID: PMC5216910 DOI: 10.18632/oncotarget.9795] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/14/2016] [Indexed: 01/05/2023] Open
Abstract
Recent studies have identified biomarkers of chronological age based on DNA methylation levels. Since active smoking contributes to a wide spectrum of aging-related diseases in adults, this study intended to examine whether active smoking exposure could accelerate the DNA methylation age in forms of age acceleration (AA, residuals of the DNA methylation age estimate regressed on chronological age). We obtained the DNA methylation profiles in whole blood samples by Illumina Infinium Human Methylation450 Beadchip array in two independent subsamples of the ESTHER study and calculated their DNA methylation ages by two recently proposed algorithms. None of the self-reported smoking indicators (smoking status, cumulative exposure and smoking cessation time) or serum cotinine levels was significantly associated with AA. On the contrary, we successfully confirmed that 66 out of 150 smoking-related CpG sites were associated with AA, even after correction for multiple testing (FDR <0.05). We further built a smoking index (SI) based on these loci and demonstrated a monotonic dose-response relationship of this index with AA. In conclusion, DNA methylation-based biological indicators for current and past smoking exposure, but not self-reported smoking information or serum cotinine levels, were found to be related to DNA methylation defined AA. Further research should address potential mechanisms underlying the observed patterns, such as potential reflections of susceptibility to environmental hazards in both smoking related methylation changes and methylation defined AA.
Collapse
Affiliation(s)
- Xu Gao
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yan Zhang
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lutz Philipp Breitling
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Pneumology and Respiratory Critical Care Medicine, Thoraxklinik, University of Heidelberg, Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
96
|
Hu R, Dai Y, Jia P, Zhao Z. ANCO-GeneDB: annotations and comprehensive analysis of candidate genes for alcohol, nicotine, cocaine and opioid dependence. Database (Oxford) 2018; 2018:5161354. [PMID: 30403795 PMCID: PMC6310508 DOI: 10.1093/database/bay121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/05/2018] [Accepted: 10/10/2018] [Indexed: 12/15/2022]
Abstract
Studies have shown that genetic factors play an important role in the risk to substance addiction and abuse. So far, various genetic and genomic studies have reported the related evidence. These rich, but highly heterogeneous, data provide us an unprecedented opportunity to systematically collect, curate and assess the genetic and genomic signals from published studies and to perform a comprehensive analysis of their features, functional roles and druggability. Such genetic data resources have been made available for other disease or phenotypes but not for major substance dependence yet. Here, we report comprehensive data collection and secondary analyses of four phenotypes of dependence: alcohol dependence, nicotine dependence, cocaine dependence and opioid dependence, collectively named as Alcohol, Nicotine, Cocaine and Opioid (ANCO) dependence. We built the ANCO-GeneDB, an ANCO-dependence-associated gene resource database. ANCO-GeneDB includes resources from genome-wide association studies and candidate gene-based studies, transcriptomic studies, methylation studies, literature mining and drug-target data, as well as the derived data such as spatial-temporal gene expression, promoters, enhancers and expression quantitative trait loci. All associated genes and genetic variants are well annotated by using the collected evidence. Based on the collected data, we performed integrative, secondary analyses to prioritize genes, pathways, eQTLs and tissues that are significantly enriched in ANCO-related phenotypes.
Collapse
Affiliation(s)
- Ruifeng Hu
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yulin Dai
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
97
|
Andersen GB, Tost J. A Summary of the Biological Processes, Disease-Associated Changes, and Clinical Applications of DNA Methylation. Methods Mol Biol 2018; 1708:3-30. [PMID: 29224136 DOI: 10.1007/978-1-4939-7481-8_1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
DNA methylation at cytosines followed by guanines, CpGs, forms one of the multiple layers of epigenetic mechanisms controlling and modulating gene expression through chromatin structure. It closely interacts with histone modifications and chromatin remodeling complexes to form the local genomic and higher-order chromatin landscape. DNA methylation is essential for proper mammalian development, crucial for imprinting and plays a role in maintaining genomic stability. DNA methylation patterns are susceptible to change in response to environmental stimuli such as diet or toxins, whereby the epigenome seems to be most vulnerable during early life. Changes of DNA methylation levels and patterns have been widely studied in several diseases, especially cancer, where interest has focused on biomarkers for early detection of cancer development, accurate diagnosis, and response to treatment, but have also been shown to occur in many other complex diseases. Recent advances in epigenome engineering technologies allow now for the large-scale assessment of the functional relevance of DNA methylation. As a stable nucleic acid-based modification that is technically easy to handle and which can be analyzed with great reproducibility and accuracy by different laboratories, DNA methylation is a promising biomarker for many applications.
Collapse
Affiliation(s)
- Gitte Brinch Andersen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Laboratory for Epigenetics & Environment, Centre National de Recherche en Génomique Humaine, CEA-Institut de Biologie Francois Jacob, Bâtiment G2, 2 rue Gaston Crémieux, 91000, Evry, France
| | - Jörg Tost
- Laboratory for Epigenetics & Environment, Centre National de Recherche en Génomique Humaine, CEA-Institut de Biologie Francois Jacob, Bâtiment G2, 2 rue Gaston Crémieux, 91000, Evry, France.
| |
Collapse
|
98
|
Abstract
Human genetic variation is a major resource in forensics, but does not allow all forensically relevant questions to be answered. Some questions may instead be addressable via epigenomics, as the epigenome acts as an interphase between the fixed genome and the dynamic environment. We envision future forensic applications of DNA methylation analysis that will broaden DNA-based forensic intelligence. Together with genetic prediction of appearance and biogeographic ancestry, epigenomic lifestyle prediction is expected to increase the ability of police to find unknown perpetrators of crime who are not identifiable using current forensic DNA profiling.
Collapse
Affiliation(s)
- Athina Vidaki
- Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Room Ee1051, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Manfred Kayser
- Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Room Ee1051, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| |
Collapse
|
99
|
Roman ÁC, Carvajal-Gonzalez JM, Merino JM, Mulero-Navarro S, Fernández-Salguero PM. The aryl hydrocarbon receptor in the crossroad of signalling networks with therapeutic value. Pharmacol Ther 2017; 185:50-63. [PMID: 29258844 DOI: 10.1016/j.pharmthera.2017.12.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is well-known for its major contributions to the cellular responses against environmental toxins and carcinogens. Notably, AhR has also emerged as a key transcription factor controlling many physiological processes including cell proliferation and apoptosis, differentiation, adhesion and migration, pluripotency and stemness. These novel functions have broadened our understanding of the signalling pathways and molecular intermediates interacting with AhR under both homeostatic and pathological conditions. Recent discoveries link AhR with the function of essential organs such as liver, skin and gonads, and with complex organismal structures including the immune and cardiovascular systems. The identification of potential endogenous ligands able to regulate AhR activity, opens the possibility of designing ad hoc molecules with pharmacological and/or therapeutic value to treat human diseases in which AhR may have a causal role. Integration of experimental data from in vitro and in vivo studies with "omic" analyses of human patients affected with cancer, immune diseases, inflammation or neurological disorders will likely contribute to validate the clinical relevance of AhR and the possible benefits of modulating its activity by pharmacologically-driven strategies. In this review, we will highlight signalling pathways involved in human diseases that could be targetable by AhR modulators and discuss the feasibility of using such molecules in therapy. The pros and cons of AhR-aimed approaches will be also mentioned.
Collapse
Affiliation(s)
- Ángel C Roman
- Champalimaud Neuroscience Programme, Champalimoud Center for the Unknown, Lisbon, Portugal
| | - José M Carvajal-Gonzalez
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
| | - Jaime M Merino
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
| | - Sonia Mulero-Navarro
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain.
| | - Pedro M Fernández-Salguero
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain.
| |
Collapse
|
100
|
Kettunen E, Hernandez-Vargas H, Cros MP, Durand G, Le Calvez-Kelm F, Stuopelyte K, Jarmalaite S, Salmenkivi K, Anttila S, Wolff H, Herceg Z, Husgafvel-Pursiainen K. Asbestos-associated genome-wide DNA methylation changes in lung cancer. Int J Cancer 2017; 141:2014-2029. [PMID: 28722770 DOI: 10.1002/ijc.30897] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/01/2017] [Accepted: 07/06/2017] [Indexed: 01/06/2023]
Abstract
Previous studies have revealed a robust association between exposure to asbestos and human lung cancer. Accumulating evidence has highlighted the role of epigenome deregulation in the mechanism of carcinogen-induced malignancies. We examined the impact of asbestos on DNA methylation. Our genome-wide studies (using Illumina HumanMethylation450K BeadChip) of lung cancer tissue and paired normal lung from 28 asbestos-exposed or non-exposed patients, mostly smokers, revealed distinctive DNA methylation changes. We identified a number of differentially methylated regions (DMR) and differentially variable, differentially methylated CpGs (DVMC), with individual CpGs further validated by pyrosequencing in an independent series of 91 non-small cell lung cancer and paired normal lung. We discovered and validated BEND4, ZSCAN31 and GPR135 as significantly hypermethylated in lung cancer. DMRs in genes such as RARB (FDR 1.1 × 10-19 , mean change in beta [Δ] -0.09), GPR135 (FDR 1.87 × 10-8 , mean Δ -0.09) and TPO (FDR 8.58 × 10-5 , mean Δ -0.11), and DVMCs in NPTN, NRG2, GLT25D2 and TRPC3 (all with p <0.05, t-test) were significantly associated with asbestos exposure status in exposed versus non-exposed lung tumors. Hypomethylation was characteristic to DVMCs in lung cancer tissue from asbestos-exposed subjects. When DVMCs related to asbestos or smoking were analyzed, 96% of the elements were unique to either of the exposures, consistent with the concept that the methylation changes in tumors may be specific for risk factors. In conclusion, we identified novel DNA methylation changes associated with lung tumors and asbestos exposure, suggesting that changes may be present in causal pathway from asbestos exposure to lung cancer.
Collapse
Affiliation(s)
- Eeva Kettunen
- Research and Service Centre for Occupational Safety, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Hector Hernandez-Vargas
- Epigenetics Group, Section of Mechanisms of Carcinogenesis, International Agency for Research on Cancer, Lyon, France
| | - Marie-Pierre Cros
- Epigenetics Group, Section of Mechanisms of Carcinogenesis, International Agency for Research on Cancer, Lyon, France
| | - Geoffroy Durand
- Genetic Cancer Susceptibility Group, Section of Genetics, International Agency for Research on Cancer, Lyon, France
| | - Florence Le Calvez-Kelm
- Genetic Cancer Susceptibility Group, Section of Genetics, International Agency for Research on Cancer, Lyon, France
| | - Kristina Stuopelyte
- Division of Human Genome Research Centre, Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Laboratory of Genetic Diagnostics, National Cancer Institute, Vilnius, Lithuania
| | - Sonata Jarmalaite
- Division of Human Genome Research Centre, Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Laboratory of Genetic Diagnostics, National Cancer Institute, Vilnius, Lithuania
| | - Kaisa Salmenkivi
- Department of Pathology, University of Helsinki, and HUSLAB, Helsinki University Hospital, Finland
| | - Sisko Anttila
- Research and Service Centre for Occupational Safety, Finnish Institute of Occupational Health, Helsinki, Finland
- Department of Pathology, University of Helsinki, and HUSLAB, Helsinki University Hospital, Finland
| | - Henrik Wolff
- Research and Service Centre for Occupational Safety, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Zdenko Herceg
- Epigenetics Group, Section of Mechanisms of Carcinogenesis, International Agency for Research on Cancer, Lyon, France
| | - Kirsti Husgafvel-Pursiainen
- Research and Service Centre for Occupational Safety, Finnish Institute of Occupational Health, Helsinki, Finland
| |
Collapse
|