1
|
Khulan B, Ye K, Shi MK, Waldman S, Marsh A, Siddiqui T, Okorozo A, Desai A, Patel D, Dobkin J, Sadoughi A, Shah C, Gera S, Peter Y, Liao W, Vijg J, Spivack SD. Normal bronchial field basal cells show persistent methylome-wide impact of tobacco smoking, including in known cancer genes. Epigenetics 2025; 20:2466382. [PMID: 39980243 PMCID: PMC11849931 DOI: 10.1080/15592294.2025.2466382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
Lung carcinogenesis is causally linked to cigarette smoking, in part by epigenetic changes. We tested whether accumulated epigenetic change in smokers is apparent in bronchial basal cells as cells of origin of squamous cell carcinoma. Using an EM-seq platform covering 53.8 million CpGs (96% of the entire genome) at an average of 7.5 sequencing reads per CpG site at a single base resolution, we evaluated cytology-normal basal cells bronchoscopically brushed from the in situ tobacco smoke-exposed 'bronchial epithelial field' and isolated by short-term primary culture from 54 human subjects. We found that mean methylation was globally lower in ever (former and current) smokers versus never smokers (p = 0.0013) across promoters, CpG shores, exons, introns, 3'-UTRs, and intergenic regions, but not in CpG islands. Among 6mers with dinucleotides flanking CpG, those containing CGCG showed no effect from smoking, while those flanked with TT and AA displayed the strongest effects. At the gene level, smoking-related differences in methylation level were observed in CDKL1, ARTN, EDC3, CYP1B1, FAM131A, and MAGI2. Among candidate cancer genes, smoking reduced the methylation level in KRAS, ROS1, CDKN1A, CHRNB4, and CADM1. We conclude that smoking reduces long-term epigenome-wide methylation in bronchial stem cells, is impacted by the flanking sequence, and persists indefinitely beyond smoking cessation.
Collapse
Affiliation(s)
- Batbayar Khulan
- Department of Pulmonary Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kenny Ye
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Miao Kevin Shi
- Department of Pulmonary Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Spencer Waldman
- Department of Pulmonary Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ava Marsh
- Department of Pulmonary Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Taha Siddiqui
- Department of Pulmonary Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aham Okorozo
- Department of Pulmonary Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aditi Desai
- Department of Pulmonary Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Dhruv Patel
- Department of Pulmonary Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jay Dobkin
- Department of Pulmonary Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ali Sadoughi
- Department of Pulmonary Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Chirag Shah
- Department of Pulmonary Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Shweta Gera
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yakov Peter
- Department of Biology, Lander College, Touro University, NY, USA
| | - Will Liao
- New York Genome Center, New York, NY, USA
| | - Jan Vijg
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Simon D. Spivack
- Department of Pulmonary Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
2
|
Jönsson J, Perfilyev A, Kugelberg U, Skog S, Lindström A, Ruhrmann S, Ofori JK, Bacos K, Rönn T, Öst A, Ling C. Impact of excess sugar on the whole genome DNA methylation pattern in human sperm. Epigenomics 2025; 17:89-104. [PMID: 39707713 PMCID: PMC11792836 DOI: 10.1080/17501911.2024.2439782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
AIMS, PATIENTS & METHODS Dietary factors may regulate the epigenome. We aimed to explore whether a diet intervention, including excess sugar, affects the methylome in human sperm, and to describe the sperm methylome. We used Whole Genome Bisulfite Sequencing (WGBS) to analyze DNA methylation in sperm taken at three time points from 15 males during a diet intervention; i) at baseline, ii) after one week on a standardized diet, and iii) after an additional week on a high-sugar diet providing 150% of their estimated total energy expenditure. RESULTS We identified seven nominal diet-associated differentially methylated regions in sperm (p < 0.05). The diet was nominally associated with methylation of 143 sites linked to fertility (e.g. AHRR, GNAS, and HDAC4), 313 sites in imprinted genes (e.g. GLIS3, PEG10, PEG3, and SNURF), and 42 sites in top 1%-expressed genes (e.g. CHD2) (p < 0.05). In sperm, 3'UTRs and introns had the highest levels of methylation, while 5'UTRs and CpG islands had the lowest levels. Non-expressed genes in human sperm were hypomethylated in exons compared with transcribed genes. CONCLUSIONS In human sperm, DNA methylation levels were linked to gene expression, and excess sugar had modest effects on methylation on imprinted and highly expressed genes, and genes affecting fertility.
Collapse
Affiliation(s)
- Josefine Jönsson
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| | - Alexander Perfilyev
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| | - Unn Kugelberg
- Department of Biomedical and Clinical Sciences, Division of Cell Biology, Linköping University, Linköping, Sweden
| | - Signe Skog
- Department of Biomedical and Clinical Sciences, Division of Cell Biology, Linköping University, Linköping, Sweden
| | - Axel Lindström
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| | - Sabrina Ruhrmann
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| | - Jones K. Ofori
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| | - Karl Bacos
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| | - Tina Rönn
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| | - Anita Öst
- Department of Biomedical and Clinical Sciences, Division of Cell Biology, Linköping University, Linköping, Sweden
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Scania University Hospital, Malmö, Sweden
| |
Collapse
|
3
|
D'Agostini F, La Maestra S. Biomarkers of genotoxic damage in pulmonary alveolar macrophages: a review. Arch Toxicol 2025; 99:211-224. [PMID: 39453481 DOI: 10.1007/s00204-024-03894-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
DNA damage is one of the primary mechanisms underlying cancer and other chronic degenerative diseases. Early evaluation of this damage in the affected cells and tissues is crucial for understanding pathogenesis and implementing effective prevention strategies. However, isolating target cells from affected organs, such as the lungs, can be challenging. Therefore, an alternative approach is to evaluate genotoxic damage in surrogate cells. Pulmonary alveolar macrophages are ideally suited for this purpose because they are in close contact with the target cells of the bronchial and alveolar epithelium, share the exact mechanisms and levels of exposure, and are easily recoverable in large numbers. This review comprehensively lists all studies using alveolar macrophages as surrogate cells to show genotoxic lung damage in humans or laboratory animals. These investigations provide fundamental information on the mechanisms of DNA damage in the lung and allow for better assessment and management of risk following exposure to inhalable genotoxic agents. Furthermore, they may be a valuable tool in cancer chemoprevention, helping the right choice of agents for clinical trials.
Collapse
Affiliation(s)
- Francesco D'Agostini
- Department of Health Sciences (DISSAL), Section of Hygiene and Preventive Medicine, University of Genoa, Via A. Pastore, 1, 16132, Genoa, Italy.
| | - Sebastiano La Maestra
- Department of Health Sciences (DISSAL), Section of Hygiene and Preventive Medicine, University of Genoa, Via A. Pastore, 1, 16132, Genoa, Italy
| |
Collapse
|
4
|
Andersen A, Beach SRH, Philibert W, Mills JA, Milefchik E, Papworth E, Dawes K, Moody J, Weeks G, Froehlich E, deBlois K, Long JD, Ahmad F, Philibert R. Heavy alcohol consumption but not smoking predicts mortality in patients with acute coronary syndrome. Epigenetics 2024; 19:2433833. [PMID: 39602337 DOI: 10.1080/15592294.2024.2433833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/26/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
The relationship of heavy alcohol consumption (HAC) and smoking to mortality in those with CHD, and mechanisms through which these effects are elicited are not clear. In order to improve our understanding, we examined the relationship of Alcohol T-Scores (ATS), an epigenetic biomarker of chronic HAC, and cg05575921 methylation, a biomarker of smoking intensity, with all-cause mortality and degree of coronary artery obstruction in a cohort of 217 subjects admitted for CHD-related acute coronary syndrome (ACS). We found that 65% of the subjects had ATS values indicative of chronic HAC. ATS values, but not cg05575921 values, were significantly associated (p < 0.02) with subsequent proband death (total of 28 deaths) with a Cox Proportional Hazards model showing a slightly larger effect of ATS levels than age on all-cause mortality survival (overall model, p < 0.003). Subjects in the highest decile of ATS scores had a 2.4-fold increase in the risk for mortality as compared to those in the lowest decile. In contrast, cg05575921 methylation (p < 0.003) but not ATS scores, were significantly inversely associated with degree of obstruction. Only 2 of the 217 subjects were referred for treatment for either smoking or drinking. We conclude that HAC is an underappreciated driver of CHD-related mortality, that those with ACS who smoke are much less likely to have significant obstruction upon cardiac imaging and that substance use treatment may be underutilized in those with CHD.
Collapse
Affiliation(s)
- Allan Andersen
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Steven R H Beach
- Center for Family Research/Department of Psychology, University of Georgia, Athens, GA, USA
| | - Willem Philibert
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - James A Mills
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Emily Milefchik
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Emma Papworth
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Kelsey Dawes
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Behavioral Diagnostics LLC, Coralville, IA, USA
| | - Joanna Moody
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Gracie Weeks
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Ellyse Froehlich
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Kaitlyn deBlois
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Jeffrey D Long
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Department of Biostatistics, University of Iowa, Iowa City, IA, USA
| | - Ferhaan Ahmad
- Department of Biostatistics, University of Iowa, Iowa City, IA, USA
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Robert Philibert
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Behavioral Diagnostics LLC, Coralville, IA, USA
| |
Collapse
|
5
|
Onuzulu CD, Lee S, Basu S, Comte J, Hai Y, Hizon N, Chadha S, Fauni MS, Halayko AJ, Pascoe CD, Jones MJ. Novel DNA methylation changes in mouse lungs associated with chronic smoking. Epigenetics 2024; 19:2322386. [PMID: 38436597 PMCID: PMC10913724 DOI: 10.1080/15592294.2024.2322386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/19/2024] [Indexed: 03/05/2024] Open
Abstract
Smoking is a potent cause of asthma exacerbations, chronic obstructive pulmonary disease (COPD) and many other health defects, and changes in DNA methylation (DNAm) have been identified as a potential link between smoking and these health outcomes. However, most studies of smoking and DNAm have been done using blood and other easily accessible tissues in humans, while evidence from more directly affected tissues such as the lungs is lacking. Here, we identified DNAm patterns in the lungs that are altered by smoking. We used an established mouse model to measure the effects of chronic smoke exposure first on lung phenotype immediately after smoking and then after a period of smoking cessation. Next, we determined whether our mouse model recapitulates previous DNAm patterns observed in smoking humans, specifically measuring DNAm at a candidate gene responsive to cigarette smoke, Cyp1a1. Finally, we carried out epigenome-wide DNAm analyses using the newly released Illumina mouse methylation microarrays. Our results recapitulate some of the phenotypes and DNAm patterns observed in human studies but reveal 32 differentially methylated genes specific to the lungs which have not been previously associated with smoking. The affected genes are associated with nicotine dependency, tumorigenesis and metastasis, immune cell dysfunction, lung function decline, and COPD. This research emphasizes the need to study CS-mediated DNAm signatures in directly affected tissues like the lungs, to fully understand mechanisms underlying CS-mediated health outcomes.
Collapse
Affiliation(s)
- Chinonye Doris Onuzulu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Samantha Lee
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sujata Basu
- Biology of Breathing Theme, Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jeannette Comte
- Biology of Breathing Theme, Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Yan Hai
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Nikho Hizon
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shivam Chadha
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Maria Shenna Fauni
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrew J. Halayko
- Biology of Breathing Theme, Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Christopher D. Pascoe
- Biology of Breathing Theme, Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Meaghan J. Jones
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
- Biology of Breathing Theme, Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
6
|
Karmaus PWF. Application of Single Cell Methods in Immunometabolism and Immunotoxicology. CURRENT OPINION IN TOXICOLOGY 2024; 39:100488. [PMID: 39091379 PMCID: PMC11290472 DOI: 10.1016/j.cotox.2024.100488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Recent developments of novel single-cell analysis techniques have rapidly advanced the fields of immunotoxicology and immunometabolism. Single-cell analyses enable the characterization of immune cells, unraveling heterogeneity, and population dynamics in response to cellular perturbations, including toxicant insults and changes in cellular metabolism. This review provides an overview of current technologies and recent discoveries, illustrating an emerging role of single-cell analyses in the field of immunotoxicology and immunometabolism. Various single-cell techniques, including flow cytometry, mass cytometry, multiplexed imaging, and sequencing, together with their applications to studying immunotoxicology and immunometabolism are discussed. This review emphasizes the potential for single-cell analyses to revolutionize our understanding of immune cell heterogeneity, uncover novel cellular therapeutic targets, and pave the way for novel mechanistic insights.
Collapse
Affiliation(s)
- Peer W F Karmaus
- National Institute of Environmental Health Sciences, US
- Institute for Integrative Toxicology, Michigan State University, US
| |
Collapse
|
7
|
Philibert R, Lei MK, Ong ML, Beach SRH. Objective Assessments of Smoking and Drinking Outperform Clinical Phenotypes in Predicting Variance in Epigenetic Aging. Genes (Basel) 2024; 15:869. [PMID: 39062648 PMCID: PMC11276345 DOI: 10.3390/genes15070869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
The reliability of the associations of the acceleration of epigenetic aging (EA) indices with clinical phenotypes other than for smoking and drinking is poorly understood. Furthermore, the majority of clinical phenotyping studies have been conducted using data from subjects of European ancestry. In order to address these limitations, we conducted clinical, physiologic, and epigenetic assessments of a cohort of 278 middle-aged African American adults and analyzed the associations with the recently described principal-components-trained version of GrimAge (i.e., PC-GrimAge) and with the DunedinPACE (PACE) index using regression analyses. We found that 74% of PC-GrimAge accelerated aging could be predicted by a simple baseline model consisting of age, sex, and methylation-sensitive digital PCR (MSdPCR) assessments of smoking and drinking. The addition of other serological, demographic, and medical history variables or PACE values did not meaningfully improve the prediction, although some variables did significantly improve the model fit. In contrast, clinical variables mapping to cardiometabolic syndrome did independently contribute to the prediction of PACE values beyond the baseline model. The PACE values were poorly correlated with the GrimAge values (r = 0.2), with little overlap in variance explained other than that conveyed by smoking and drinking. The results suggest that EA indices may differ in the clinical information that they provide and may have significant limitations as screening tools to guide patient care.
Collapse
Affiliation(s)
- Robert Philibert
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242, USA
- Behavioral Diagnostics LLC, Coralville, IA 52241, USA
| | - Man-Kit Lei
- Department of Sociology, University of Georgia, Athens, GA 30602, USA;
- Center for Family Research, University of Georgia, Athens, GA 30602, USA; (M.L.O.); (S.R.H.B.)
| | - Mei Ling Ong
- Center for Family Research, University of Georgia, Athens, GA 30602, USA; (M.L.O.); (S.R.H.B.)
| | - Steven R. H. Beach
- Center for Family Research, University of Georgia, Athens, GA 30602, USA; (M.L.O.); (S.R.H.B.)
- Department of Psychology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
8
|
Li JL, Jain N, Tamayo LI, Tong L, Jasmine F, Kibriya MG, Demanelis K, Oliva M, Chen LS, Pierce BL. The association of cigarette smoking with DNA methylation and gene expression in human tissue samples. Am J Hum Genet 2024; 111:636-653. [PMID: 38490207 PMCID: PMC11023923 DOI: 10.1016/j.ajhg.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 02/17/2024] [Accepted: 02/21/2024] [Indexed: 03/17/2024] Open
Abstract
Cigarette smoking adversely affects many aspects of human health, and epigenetic responses to smoking may reflect mechanisms that mediate or defend against these effects. Prior studies of smoking and DNA methylation (DNAm), typically measured in leukocytes, have identified numerous smoking-associated regions (e.g., AHRR). To identify smoking-associated DNAm features in typically inaccessible tissues, we generated array-based DNAm data for 916 tissue samples from the GTEx (Genotype-Tissue Expression) project representing 9 tissue types (lung, colon, ovary, prostate, blood, breast, testis, kidney, and muscle). We identified 6,350 smoking-associated CpGs in lung tissue (n = 212) and 2,735 in colon tissue (n = 210), most not reported previously. For all 7 other tissue types (sample sizes 38-153), no clear associations were observed (false discovery rate 0.05), but some tissues showed enrichment for smoking-associated CpGs reported previously. For 1,646 loci (in lung) and 22 (in colon), smoking was associated with both DNAm and local gene expression. For loci detected in both lung and colon (e.g., AHRR, CYP1B1, CYP1A1), top CpGs often differed between tissues, but similar clusters of hyper- or hypomethylated CpGs were observed, with hypomethylation at regulatory elements corresponding to increased expression. For lung tissue, 17 hallmark gene sets were enriched for smoking-associated CpGs, including xenobiotic- and cancer-related gene sets. At least four smoking-associated regions in lung were impacted by lung methylation quantitative trait loci (QTLs) that co-localize with genome-wide association study (GWAS) signals for lung function (FEV1/FVC), suggesting epigenetic alterations can mediate the effects of smoking on lung health. Our multi-tissue approach has identified smoking-associated regions in disease-relevant tissues, including effects that are shared across tissue types.
Collapse
Affiliation(s)
- James L Li
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA; Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL 60637, USA
| | - Niyati Jain
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA; Committee on Genetics, Genomics, Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Lizeth I Tamayo
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Lin Tong
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Farzana Jasmine
- Institute for Population and Precision Health (IPPH), Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA
| | - Muhammad G Kibriya
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Kathryn Demanelis
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Meritxell Oliva
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA; Genomics Research Center, AbbVie, North Chicago, IL 60064, USA
| | - Lin S Chen
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Brandon L Pierce
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA; Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA; Comprehensive Cancer Center, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
9
|
Garrett ME, Dennis MF, Bourassa KJ, Hauser MA, Kimbrel NA, Beckham JC, Ashley-Koch AE. Genome-wide DNA methylation analysis of cannabis use disorder in a veteran cohort enriched for posttraumatic stress disorder. Psychiatry Res 2024; 333:115757. [PMID: 38309009 PMCID: PMC10922626 DOI: 10.1016/j.psychres.2024.115757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/05/2024]
Abstract
Cannabis use has been increasing over the past decade, not only in the general US population, but particularly among military veterans. With this rise in use has come a concomitant increase in cannabis use disorder (CUD) among veterans. Here, we performed an epigenome-wide association study for lifetime CUD in an Iraq/Afghanistan era veteran cohort enriched for posttraumatic stress disorder (PTSD) comprising 2,310 total subjects (1,109 non-Hispanic black and 1,201 non-Hispanic white). We also investigated CUD interactions with current PTSD status and examined potential indirect effects of DNA methylation (DNAm) on the relationship between CUD and psychiatric diagnoses. Four CpGs were associated with lifetime CUD, even after controlling for the effects of current smoking (AHRR cg05575921, LINC00299 cg23079012, VWA7 cg22112841, and FAM70A cg08760398). Importantly, cg05575921, a CpG strongly linked to smoking, remained associated with lifetime CUD even when restricting the analysis to veterans who reported never smoking cigarettes. Moreover, CUD interacted with current PTSD to affect cg05575921 and cg23079012 such that those with both CUD and PTSD displayed significantly lower DNAm compared to the other groups. Finally, we provide preliminary evidence that AHRR cg05575921 helps explain the association between CUD and any psychiatric diagnoses, specifically mood disorders.
Collapse
Affiliation(s)
- Melanie E Garrett
- Duke Molecular Physiology Institute, Duke University Medical Center, 300N Duke St, Durham, NC 27701, USA
| | - Michelle F Dennis
- Durham Veterans Affairs (VA) Health Care System, Durham, NC, USA; VA Mid-Atlantic Mental Illness Research, Education, and Clinical Center, Durham, NC, USA; Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Kyle J Bourassa
- Durham Veterans Affairs (VA) Health Care System, Durham, NC, USA; VA Mid-Atlantic Mental Illness Research, Education, and Clinical Center, Durham, NC, USA; Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC, USA
| | - Michael A Hauser
- Duke Molecular Physiology Institute, Duke University Medical Center, 300N Duke St, Durham, NC 27701, USA
| | - Nathan A Kimbrel
- Durham Veterans Affairs (VA) Health Care System, Durham, NC, USA; VA Mid-Atlantic Mental Illness Research, Education, and Clinical Center, Durham, NC, USA; Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Jean C Beckham
- Durham Veterans Affairs (VA) Health Care System, Durham, NC, USA; VA Mid-Atlantic Mental Illness Research, Education, and Clinical Center, Durham, NC, USA; Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Allison E Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, 300N Duke St, Durham, NC 27701, USA.
| |
Collapse
|
10
|
Awada Z, Cahais V, Cuenin C, Akika R, Silva Almeida Vicente AL, Makki M, Tamim H, Herceg Z, Khoueiry Zgheib N, Ghantous A. Waterpipe and cigarette epigenome analysis reveals markers implicated in addiction and smoking type inference. ENVIRONMENT INTERNATIONAL 2023; 182:108260. [PMID: 38006773 PMCID: PMC10716859 DOI: 10.1016/j.envint.2023.108260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/02/2023] [Accepted: 10/09/2023] [Indexed: 11/27/2023]
Abstract
Waterpipe smoking is frequent in the Middle East and Africa with emerging prevalence worldwide. The epigenome acts as a molecular sensor to exposures and a crucial driver in several diseases. With the widespread use of waterpipe smoking, it is timely to investigate its epigenomic markers and their role in addiction, as a central player in disease prevention and therapeutic strategies. DNA methylome-wide profiling was performed on an exposure-rich population from the Middle East, constituting of 216 blood samples split equally between never, cigarette-only and waterpipe-only smokers. Waterpipe smokers showed predominantly distinct epigenetic markers from cigarette smokers, even though both smoking forms are tobacco-based. Moreover, each smoking form could be accurately (∼90 %) inferred from the DNA methylome using machine learning. Top markers showed dose-response relationship with extent of smoking and were validated using independent technologies and additional samples (total N = 284). Smoking markers were enriched in regulatory regions and several biological pathways, primarily addiction. The epigenetically altered genes were not associated with genetic etiology of tobacco use, and the methylation levels of addiction genes, in particular, were more likely to reverse after smoking cessation. In contrast, other epigenetic markers continued to feature smoking exposure after cessation, which may explain long-term health effects observed in former smokers. This study reports, for the first time, blood epigenome-wide markers of waterpipe smokers and reveals new markers of cigarette smoking, with implications in mechanisms of addiction and the capacity to discriminate between different smoking types. These markers may offer actionable targets to reverse the epigenetic memory of addiction and can guide future prevention strategies for tobacco smoking as the most preventable cause of illnesses worldwide.
Collapse
Affiliation(s)
- Zainab Awada
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Vincent Cahais
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Cyrille Cuenin
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Reem Akika
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Anna Luiza Silva Almeida Vicente
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France; Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Maha Makki
- Clinical Research Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Hani Tamim
- Clinical Research Institute, American University of Beirut Medical Center, Beirut, Lebanon; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Zdenko Herceg
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France
| | - Nathalie Khoueiry Zgheib
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| | - Akram Ghantous
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, Lyon, France.
| |
Collapse
|
11
|
Carreras-Gallo N, Dwaraka VB, Cáceres A, Smith R, Mendez TL, Went H, Gonzalez JR. Impact of tobacco, alcohol, and marijuana on genome-wide DNA methylation and its relationship with hypertension. Epigenetics 2023; 18:2214392. [PMID: 37216580 DOI: 10.1080/15592294.2023.2214392] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 04/13/2023] [Accepted: 05/09/2023] [Indexed: 05/24/2023] Open
Abstract
Tobacco, alcohol, and marijuana consumption is an important public health problem because of their high use worldwide and their association with the risk of mortality and many health conditions, such as hypertension, which is the commonest risk factor for death throughout the world. A likely pathway of action of substance consumption leading to persistent hypertension is DNA methylation. Here, we evaluated the effects of tobacco, alcohol, and marijuana on DNA methylation in the same cohort (N = 3,424). Three epigenome-wide association studies (EWAS) were assessed in whole blood using the InfiniumHumanMethylationEPIC BeadChip. We also evaluated the mediation of the top CpG sites in the association between substance consumption and hypertension. Our analyses showed 2,569 CpG sites differentially methylated by alcohol drinking and 528 by tobacco smoking. We did not find significant associations with marijuana consumption after correcting for multiple comparisons. We found 61 genes overlapping between alcohol and tobacco that were enriched in biological processes involved in the nervous and cardiovascular systems. In the mediation analysis, we found 66 CpG sites that significantly mediated the effect of alcohol consumption on hypertension. The top alcohol-related CpG site (cg06690548, P-value = 5.9·10-83) mapped to SLC7A11 strongly mediated 70.5% of the effect of alcohol consumption on hypertension (P-value = 0.006). Our findings suggest that DNA methylation should be considered for new targets in hypertension prevention and management, particularly concerning alcohol consumption. Our data also encourage further research into the use of methylation in blood to study the neurological and cardiovascular effects of substance consumption.
Collapse
Affiliation(s)
| | | | - Alejandro Cáceres
- Epidemiology, Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Mathematics, Escola d'Enginyeria de Barcelona Est (EEBE), Universitat Politècnica de Catalunya, Barcelona, Spain
| | | | | | | | - Juan R Gonzalez
- Epidemiology, Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Mathematics, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
12
|
Lu Y, Deng M, Yin Y, Hou G, Zhou X. Global Trends in Research Regarding Macrophages Associated with Chronic Obstructive Pulmonary Disease: A Bibliometric Analysis from 2011 to 2022. Int J Chron Obstruct Pulmon Dis 2023; 18:2163-2177. [PMID: 37810373 PMCID: PMC10558051 DOI: 10.2147/copd.s419634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/21/2023] [Indexed: 10/10/2023] Open
Abstract
Purpose Chronic obstructive pulmonary disease (COPD) is a prevalent respiratory condition characterized by chronic airway inflammation, where macrophages from the innate immune system may exert a pivotal influence. Our study aimed to summarize the present state of knowledge and to identify the focal points and emerging developments regarding macrophages associated with COPD through bibliometrics. Methods Publications regarding research on macrophages associated with COPD from January 1, 2011, to January 1, 2022, were retrieved from the Science Citation Index-Expanded (SCI-E) which is part of the Web of Science database. In total, 1521 publications were analyzed using bibliometric methodology. VOSviewer was used to analyze the annual publications, countries, institutions, authors, journals, and research hotspots. Results Based on the bibliometric analysis, publications relating to macrophages associated with COPD tended to increase from 2011 to 2022. The United States was the largest producer and most influential country in this field. Research during the past decade has focused on inflammation in the lungs. Most previous studies have mainly focused on the mechanisms that promote the initiation and progression of COPD. Macrophage-related oxidative stress and immunity, communication between macrophages and epithelial cells, and interventions for acute exacerbations have become the focus of more recent studies and will become a hot topic in the future. Conclusion Global research on macrophage-associated COPD has been growing rapidly in the past decade. The hot topics in this field gradually tended to shift focus from "inflammation" to "oxidative stress", "epithelial-cells", and "exacerbations". The significance of macrophages in coordinating immune responses, interacting with other cells, and exhibiting dysregulated capacities has attracted increasing attention to COPD pathogenesis. The adoption of new technologies may provide a more promising and comprehensive understanding of the specific role of macrophages in COPD in the future.
Collapse
Affiliation(s)
- Ye Lu
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Mingming Deng
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine, Beijing, People’s Republic of China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- National Clinical Research Center for Respiratory Diseases, Beijing, People’s Republic of China
| | - Yan Yin
- Department of Respiratory and Critical Care Medicine, First Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Gang Hou
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine, Beijing, People’s Republic of China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- National Clinical Research Center for Respiratory Diseases, Beijing, People’s Republic of China
| | - Xiaoming Zhou
- Department of Respirology, Fuwai Hospital, National Center for Cardiovascular Diseases, Beijing, People’s Republic of China
| |
Collapse
|
13
|
Broyles D, Philibert R. Precision epigenetics provides a scalable pathway for improving coronary heart disease care globally. Epigenomics 2023; 15:805-818. [PMID: 37702023 DOI: 10.2217/epi-2023-0233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023] Open
Abstract
Coronary heart disease (CHD) is the world's leading cause of death. Up to 90% of all CHD deaths are preventable, but effective prevention of this mortality requires more scalable, precise methods for assessing CHD status and monitoring treatment response. Unfortunately, current diagnostic methods have barriers to implementation, particularly in rural areas and lower-income countries. This gap may be bridged by highly scalable advances in DNA methylation testing methods and artificial intelligence. Herein, we review prior studies of CHD related to methylation alone and in combination with other biovariables. We compare these new methods with established methods for diagnosing CHD. Finally, we outline pathways through which methylation-based testing methods may allow the democratization of improved methods for assessing CHD globally.
Collapse
Affiliation(s)
- Damon Broyles
- Mercy Technology Services, St. Louis, MO 63127, USA
- Mercy Precision Medicine, Chesterfield, MO 63017, USA
| | - Robert Philibert
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242, USA
- Cardio Diagnostics Inc, Chicago, IL 60642, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
14
|
Bosch AJT, Rohm TV, AlAsfoor S, Low AJY, Baumann Z, Parayil N, Noreen F, Roux J, Meier DT, Cavelti-Weder C. Diesel Exhaust Particle (DEP)-induced glucose intolerance is driven by an intestinal innate immune response and NLRP3 activation in mice. Part Fibre Toxicol 2023; 20:25. [PMID: 37400850 DOI: 10.1186/s12989-023-00536-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/20/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND We previously found that air pollution particles reaching the gastrointestinal tract elicit gut inflammation as shown by up-regulated gene expression of pro-inflammatory cytokines and monocyte/macrophage markers. This inflammatory response was associated with beta-cell dysfunction and glucose intolerance. So far, it remains unclear whether gut inflammatory changes upon oral air pollution exposure are causally linked to the development of diabetes. Hence, our aim was to assess the role of immune cells in mediating glucose intolerance instigated by orally administered air pollutants. METHODS To assess immune-mediated mechanisms underlying air pollution-induced glucose intolerance, we administered diesel exhaust particles (DEP; NIST 1650b, 12 µg five days/week) or phosphate-buffered saline (PBS) via gavage for up to 10 months to wild-type mice and mice with genetic or pharmacological depletion of innate or adaptive immune cells. We performed unbiased RNA-sequencing of intestinal macrophages to elucidate signaling pathways that could be pharmacologically targeted and applied an in vitro approach to confirm these pathways. RESULTS Oral exposure to air pollution particles induced an interferon and inflammatory signature in colon macrophages together with a decrease of CCR2- anti-inflammatory/resident macrophages. Depletion of macrophages, NLRP3 or IL-1β protected mice from air pollution-induced glucose intolerance. On the contrary, Rag2-/- mice lacking adaptive immune cells developed pronounced gut inflammation and glucose intolerance upon oral DEP exposure. CONCLUSION In mice, oral exposure to air pollution particles triggers an immune-mediated response in intestinal macrophages that contributes to the development of a diabetes-like phenotype. These findings point towards new pharmacologic targets in diabetes instigated by air pollution particles.
Collapse
Affiliation(s)
- Angela J T Bosch
- Department of Biomedicine, University of Basel, Basel, 4031, Switzerland
| | - Theresa V Rohm
- Department of Biomedicine, University of Basel, Basel, 4031, Switzerland
| | - Shefaa AlAsfoor
- Department of Biomedicine, University of Basel, Basel, 4031, Switzerland
| | - Andy J Y Low
- Department of Biomedicine, University of Basel, Basel, 4031, Switzerland
| | - Zora Baumann
- Department of Biomedicine, University of Basel, Basel, 4031, Switzerland
| | - Neena Parayil
- Department of Biomedicine, University of Basel, Basel, 4031, Switzerland
| | - Faiza Noreen
- Department of Biomedicine, University of Basel, Basel, 4031, Switzerland
- Swiss Institute of Bioinformatics, Basel, 4031, Switzerland
| | - Julien Roux
- Department of Biomedicine, University of Basel, Basel, 4031, Switzerland
- Swiss Institute of Bioinformatics, Basel, 4031, Switzerland
| | - Daniel T Meier
- Department of Biomedicine, University of Basel, Basel, 4031, Switzerland
| | - Claudia Cavelti-Weder
- Department of Biomedicine, University of Basel, Basel, 4031, Switzerland.
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, 4031, Switzerland.
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Zurich, Switzerland.
- University Hospital Zurich, Rämistrasse 100, Zürich, 8009, Switzerland.
| |
Collapse
|
15
|
Nannini DR, Zheng Y, Joyce BT, Kim K, Gao T, Wang J, Jacobs DR, Schreiner PJ, Yaffe K, Greenland P, Lloyd-Jones DM, Hou L. Genome-wide DNA methylation association study of recent and cumulative marijuana use in middle aged adults. Mol Psychiatry 2023; 28:2572-2582. [PMID: 37258616 PMCID: PMC10611566 DOI: 10.1038/s41380-023-02106-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/24/2023] [Accepted: 05/03/2023] [Indexed: 06/02/2023]
Abstract
Marijuana is a widely used psychoactive substance in the US and medical and recreational legalization has risen over the past decade. Despite the growing number of individuals using marijuana, studies investigating the association between epigenetic factors and recent and cumulative marijuana use remain limited. We therefore investigated the association between recent and cumulative marijuana use and DNA methylation levels. Participants from the Coronary Artery Risk Development in Young Adults Study with whole blood collected at examination years (Y) 15 and Y20 were randomly selected to undergo DNA methylation profiling at both timepoints using the Illumina MethylationEPIC BeadChip. Recent use of marijuana was queried at each examination and used to estimate cumulative marijuana use from Y0 to Y15 and Y20. At Y15 (n = 1023), we observed 22 and 31 methylation markers associated (FDR P ≤ 0.05) with recent and cumulative marijuana use and 132 and 16 methylation markers at Y20 (n = 883), respectively. We replicated 8 previously reported methylation markers associated with marijuana use. We further identified 640 cis-meQTLs and 198 DMRs associated with recent and cumulative use at Y15 and Y20. Differentially methylated genes were statistically overrepresented in pathways relating to cellular proliferation, hormone signaling, and infections as well as schizophrenia, bipolar disorder, and substance-related disorders. We identified numerous methylation markers, pathways, and diseases associated with recent and cumulative marijuana use in middle-aged adults, providing additional insight into the association between marijuana use and the epigenome. These results provide novel insights into the role marijuana has on the epigenome and related health conditions.
Collapse
Affiliation(s)
- Drew R Nannini
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Yinan Zheng
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Brian T Joyce
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kyeezu Kim
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tao Gao
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jun Wang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - David R Jacobs
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Pamela J Schreiner
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Kristine Yaffe
- University of California at San Francisco School of Medicine, San Francisco, CA, USA
| | - Philip Greenland
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Donald M Lloyd-Jones
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
16
|
Skov-Jeppesen SM, Kobylecki CJ, Jacobsen KK, Bojesen SE. Changing Smoking Behavior and Epigenetics: A Longitudinal Study of 4,432 Individuals From the General Population. Chest 2023; 163:1565-1575. [PMID: 36621758 PMCID: PMC10258440 DOI: 10.1016/j.chest.2022.12.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/07/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Hypomethylation of the aryl hydrocarbon receptor repressor (AHRR) gene indicates long-term smoking exposure and might therefore be a monitor for smoking-induced disease risk. However, studies of individual longitudinal changes in AHRR methylation are sparse. RESEARCH QUESTION How does the recovery of AHRR methylation depend on change in smoking behaviors and demographic variables? STUDY DESIGN AND METHODS This study included 4,432 individuals from the Copenhagen City Heart Study, with baseline and follow-up blood samples and smoking information collected approximately 10 years apart. AHRR methylation at the cg05575921 site was measured in bisulfite-treated leukocyte DNA. Four smoking groups were defined: participants who never smoked (Never-Never), participants who formerly smoked (Former-Former), participants who quit during the study period (Current-Former), and individuals who smoked at both baseline and follow-up (Current-Current). Methylation recovery was defined as the increase in AHRR methylation between baseline and follow-up examination. RESULTS Methylation recovery was highest among participants who quit, with a median methylation recovery of 5.58% (interquartile range, 1.79; 9.15) vs 1.64% (interquartile range, -1.88; 4.96) in the Current-Current group (P < .0001). In individuals who quit smoking, older age was associated with lower methylation recovery (P < .0001). In participants who quit aged > 65 years, methylation recovery was 5.9% at 5.6 years after quitting; methylation recovery was 8.5% after 2.8 years for participants who quit aged < 55 years. INTERPRETATION AHRR methylation recovered after individuals quit smoking, and recovery was more pronounced and occurred faster in younger compared with older interim quitters.
Collapse
Affiliation(s)
- Sune Moeller Skov-Jeppesen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Camilla Jannie Kobylecki
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Katja Kemp Jacobsen
- Department of Technology, Faculty of Health and Technology, University College Copenhagen, Copenhagen, Denmark
| | - Stig Egil Bojesen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark; The Copenhagen City Heart Study, Copenhagen University Hospital, Frederiksberg and Bispebjerg Hospital, Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
17
|
Philibert R, Hoffman RM. Assessing Our Will to Change. Chest 2023; 163:1360-1361. [PMID: 37295878 DOI: 10.1016/j.chest.2023.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 06/12/2023] Open
Affiliation(s)
- Robert Philibert
- Department of Psychiatry, University of Iowa, Iowa City, IA; Behavioral Diagnostics LLC, Coralville, IA.
| | | |
Collapse
|
18
|
Zailani H, Satyanarayanan SK, Liao WC, Liao HF, Huang SY, Gałecki P, Su KP, Chang JPC. Omega-3 Polyunsaturated Fatty Acids in Managing Comorbid Mood Disorders in Chronic Obstructive Pulmonary Disease (COPD): A Review. J Clin Med 2023; 12:jcm12072653. [PMID: 37048736 PMCID: PMC10095486 DOI: 10.3390/jcm12072653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/15/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the third-leading cause of mortality globally, significantly affecting people over 40 years old. COPD is often comorbid with mood disorders; however, they are frequently neglected or undiagnosed in COPD management, thus resulting in unintended treatment outcomes and higher mortality associated with the disease. Although the exact link between COPD and mood disorders remains to be ascertained, there is a broader opinion that inflammatory reactions in the lungs, blood, and inflammation-induced changes in the brain could orchestrate the onset of mood disorders in COPD. Although the current management of mood disorders such as depression in COPD involves using antidepressants, their use has been limited due to tolerability issues. On the other hand, as omega-3 polyunsaturated fatty acids (n-3 PUFAs) play a vital role in regulating inflammatory responses, they could be promising alternatives in managing mood disorders in COPD. This review discusses comorbid mood disorders in COPD as well as their influence on the progression and management of COPD. The underlying mechanisms of comorbid mood disorders in COPD will also be discussed, along with the potential role of n-3 PUFAs in managing these conditions.
Collapse
Affiliation(s)
- Halliru Zailani
- Mind-Body Interface Laboratory (MBI-Lab), Department of Psychiatry, China Medical University Hospital, Taichung 404, Taiwan
- Graduate Institute of Nutrition, China Medical University, Taichung 404, Taiwan
| | - Senthil Kumaran Satyanarayanan
- Mind-Body Interface Laboratory (MBI-Lab), Department of Psychiatry, China Medical University Hospital, Taichung 404, Taiwan
| | - Wei-Chih Liao
- Division of Pulmonary and Critical Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Hsien-Feng Liao
- Mind-Body Interface Laboratory (MBI-Lab), Department of Psychiatry, China Medical University Hospital, Taichung 404, Taiwan
| | - Shih-Yi Huang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110, Taiwan
- Nutrition Research Centre, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Piotr Gałecki
- Department of Adult Psychiatry, Medical University of Lodz, 91-229 Lodz, Poland
| | - Kuan-Pin Su
- Mind-Body Interface Laboratory (MBI-Lab), Department of Psychiatry, China Medical University Hospital, Taichung 404, Taiwan
- College of Medicine, China Medical University, Taichung 404, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
- An-Nan Hospital, China Medical University, Tainan 833, Taiwan
| | - Jane Pei-Chen Chang
- Mind-Body Interface Laboratory (MBI-Lab), Department of Psychiatry, China Medical University Hospital, Taichung 404, Taiwan
- College of Medicine, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
19
|
Kurihara C, Kuniyoshi KM, Rehan VK. Preterm Birth, Developmental Smoke/Nicotine Exposure, and Life-Long Pulmonary Sequelae. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10040608. [PMID: 37189857 DOI: 10.3390/children10040608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 05/17/2023]
Abstract
This review delineates the main pulmonary issues related to preterm birth, perinatal tobacco/nicotine exposure, and its effects on offspring, focusing on respiratory health and its possible transmission to subsequent generations. We review the extent of the problem of preterm birth, prematurity-related pulmonary effects, and the associated increased risk of asthma later in life. We then review the impact of developmental tobacco/nicotine exposure on offspring asthma and the significance of transgenerational pulmonary effects following perinatal tobacco/nicotine exposure, possibly via its effects on germline epigenetics.
Collapse
Affiliation(s)
- Chie Kurihara
- Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Katherine M Kuniyoshi
- Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Virender K Rehan
- Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
20
|
Ghemrawi M, Tejero NF, Duncan G, McCord B. Pyrosequencing: Current forensic methodology and future applications-a review. Electrophoresis 2023; 44:298-312. [PMID: 36168852 DOI: 10.1002/elps.202200177] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 02/01/2023]
Abstract
The recent development of small, single-amplicon-based benchtop systems for pyrosequencing has opened up a host of novel procedures for applications in forensic science. Pyrosequencing is a sequencing by synthesis technique, based on chemiluminescent inorganic pyrophosphate detection. This review explains the pyrosequencing workflow and illustrates the step-by-step chemistry, followed by a description of the assay design and factors to keep in mind for an exemplary assay. Existing and potential forensic applications are highlighted using this technology. Current applications include identifying species, identifying bodily fluids, and determining smoking status. We also review progress in potential applications for the future, including research on distinguishing monozygotic twins, detecting alcohol and drug abuse, and other phenotypic characteristics such as diet and body mass index. Overall, the versatility of the pyrosequencing technologies renders it a useful tool in forensic genomics.
Collapse
Affiliation(s)
- Mirna Ghemrawi
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
| | - Nicole Fernandez Tejero
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
| | - George Duncan
- Halmos College of Natural Sciences and Oceanography, Nova Southeastern University, Dania Beach, Florida, USA
| | - Bruce McCord
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida, USA
| |
Collapse
|
21
|
Long JD, Gehlsen MP, Moody J, Weeks G, Philibert R. Predictors of Smoking in Older Adults and an Epigenetic Validation of Self-Report. Genes (Basel) 2022; 14:genes14010025. [PMID: 36672765 PMCID: PMC9912258 DOI: 10.3390/genes14010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022] Open
Abstract
There are several established predictors of smoking, but it is unknown if these predictors operate similarly for young and old smokers. We examined clinical data from the National Lung Screening Trial (NLST) to determine the predictive ability of gender, body mass index (BMI), marital status, and race on smoking behavior, with emphasis on gender interactions. In addition, we validated the self-report of smoking behaviors for a subgroup that had available epigenetic data in the form of cg05575921 methylation. Participants were N=9572 current or former smokers from the NLST biofluids database, age 55-74, minimum of 30 pack years, and mostly White. A subgroup of N=3084 who had DNA were used for the self-report validation analysis. The predictor analysis was based on the larger group and used penalized logistic regression to predict the self-report of being a former or current smoker at baseline. Cg05575921 methylation showed a moderate ability to discriminate among former and current smokers, AUC = 0.85 (95% confidence interval = [0.83, 0.86]). The final selected variables for the prediction model were BMI, gender, BMI by gender, age, divorced (vs. married), education, and race. The gender by BMI interaction was such that males had a higher probability of current smoking for lower BMI, but this switched to females having higher current smoking for overweight to obese. There is evidence that the self-reported smoking behavior in NLST is moderately accurate. The results of the primary analysis are consistent with the general smoking literature, and our results provide additional specificity regarding the gender by BMI interaction. Body weight issues might play a role in smoking cessation for older established smokers in a similar manner as younger smokers. It could be that women have less success with cessation when their BMI increases.
Collapse
Affiliation(s)
- Jeffrey D. Long
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa, IA 52242, USA; (J.D.L.); (J.M.); (G.W.)
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa, IA 52242, USA
| | - Michael P. Gehlsen
- South Saint Paul Public Schools, 104 5th Ave. S, South Saint Paul, MN 55075, USA;
| | - Joanna Moody
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa, IA 52242, USA; (J.D.L.); (J.M.); (G.W.)
| | - Gracie Weeks
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa, IA 52242, USA; (J.D.L.); (J.M.); (G.W.)
| | - Robert Philibert
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa, IA 52242, USA; (J.D.L.); (J.M.); (G.W.)
- Behavioral Diagnostics LLC, 2500 Crosspark Rd., Suite W245, Coralville, IA 53341, USA
- Department of Biomedical Engineering, University of Iowa, Iowa, IA 52242, USA
- Correspondence:
| |
Collapse
|
22
|
Philibert R, Dawes K, Moody J, Hoffman R, Sieren J, Long J. Using Cg05575921 methylation to predict lung cancer risk: a potentially bias-free precision epigenetics approach. Epigenetics 2022; 17:2096-2108. [PMID: 35920547 PMCID: PMC9665144 DOI: 10.1080/15592294.2022.2108082] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 07/25/2022] [Indexed: 01/25/2023] Open
Abstract
The decision to engage in lung cancer screening (LCS) necessitates weighing benefits versus harms. Previously, clinicians in the United States have used the PLCOM2012 algorithm to guide LCS decision-making. However, that formula contains race and gender-based variables. Previously, using data from a European study, Bojesen and colleagues have suggested that cg05575921 methylation could guide decision-making. To test this hypothesis in a more diverse American population, we examined DNA and clinical data from 3081 subjects from the National Lung Screening Trial (NLST) study. Using survival analysis, we found a simple linear predictor consisting of age, pack-year consumption and cg05575921, to have the best predictive power among several alternatives (AUC = 0.66). Results showed that the highest quartile of risk was more than 2-fold more likely to develop lung cancer than those in the lowest quartile. Race, ethnicity, and gender had no effect on prediction with both cg05575921 and pack years contributing equally (both p < 0.003) to risk prediction. Current smokers had considerably lower methylation than former smokers (46% vs 67%; p < 0.001) with the average methylation of those who quit approaching 80% after 25 years of cessation. Finally, current male smokers had lower mean cg05575921 percentage than female smokers (46% vs 49%; p < 0.001). We conclude that cg05575921 (along with age and pack years) can be used to guide LCS decision-making, and additional studies might focus on how best to use methylation to inform decision-making.
Collapse
Affiliation(s)
- Rob Philibert
- Behavioural Diagnostics LLC, Coralville, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Kelsey Dawes
- Behavioural Diagnostics LLC, Coralville, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Joanna Moody
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Richard Hoffman
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Jessica Sieren
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
- Department of Radiology, University of Iowa, Iowa City, IA, USA
- Department of Biostatistics, University of Iowa, Iowa City, IA, USA
| | - Jeffrey Long
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
- Department of Biostatistics, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
23
|
Chen Q, Nwozor KO, van den Berge M, Slebos DJ, Faiz A, Jonker MR, Boezen HM, Heijink IH, de Vries M. From Differential DNA Methylation in COPD to Mitochondria: Regulation of AHRR Expression Affects Airway Epithelial Response to Cigarette Smoke. Cells 2022; 11:3423. [PMID: 36359818 PMCID: PMC9656229 DOI: 10.3390/cells11213423] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 08/01/2023] Open
Abstract
Cigarette smoking causes hypomethylation of the gene Aryl Hydrocarbon Receptor Repressor (AHRR), which regulates detoxification and oxidative stress-responses. We investigated whether AHRR DNA methylation is related to chronic obstructive pulmonary disease (COPD) and studied its function in airway epithelial cells (AECs). The association with COPD was assessed in blood from never and current smokers with/without COPD, and in AECs from ex-smoking non-COPD controls and GOLD stage II-IV COPD patients cultured with/without cigarette smoke extract (CSE). The effect of CRISPR/Cas9-induced AHRR knockout on proliferation, CSE-induced mitochondrial membrane potential and apoptosis/necrosis in human bronchial epithelial 16HBE cells was studied. In blood, DNA methylation of AHRR at cg05575921 and cg21161138 was lower in smoking COPD subjects than smoking controls. In vitro, AHRR DNA methylation at these CpG-sites was lower in COPD-derived than control-derived AECs only upon CSE exposure. Upon AHRR knockout, we found a lower proliferation rate at baseline, stronger CSE-induced decrease in mitochondrial membrane potential, and higher CSE-induced late apoptosis/necroptosis. Together, our results show lower DNA methylation of AHRR upon smoking in COPD patients compared to non-COPD controls. Our data suggest that higher airway epithelial AHRR expression may lead to impaired cigarette smoke-induced mitochondrial dysfunction and apoptosis/necroptosis, potentially promoting unprogrammed/immunogenic cell death.
Collapse
Affiliation(s)
- Qing Chen
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, 9713 GZ Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), 9713 GZ Groningen, The Netherlands
| | - Kingsley Okechukwu Nwozor
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, 9713 GZ Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), 9713 GZ Groningen, The Netherlands
- Centre for Heart Lung Innovation, Department of Anesthesiology, Pharmacology & Therapeutics, The University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Maarten van den Berge
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), 9713 GZ Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology Disease, 9713 GZ Groningen, The Netherlands
| | - Dirk-Jan Slebos
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), 9713 GZ Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology Disease, 9713 GZ Groningen, The Netherlands
| | - Alen Faiz
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), 9713 GZ Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology Disease, 9713 GZ Groningen, The Netherlands
- Respiratory Bioinformatics and Molecular Biology (RBMB), School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Marnix R. Jonker
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, 9713 GZ Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), 9713 GZ Groningen, The Netherlands
| | - H. Marike Boezen
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), 9713 GZ Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, 9713 GZ Groningen, The Netherlands
| | - Irene H. Heijink
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, 9713 GZ Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), 9713 GZ Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pulmonology Disease, 9713 GZ Groningen, The Netherlands
| | - Maaike de Vries
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), 9713 GZ Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
24
|
Role of Long Noncoding RNAs in Smoking-Induced Lung Cancer: An In Silico Study. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7169353. [PMID: 35529255 PMCID: PMC9070410 DOI: 10.1155/2022/7169353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/13/2022] [Accepted: 04/05/2022] [Indexed: 11/17/2022]
Abstract
The prevalence of lung cancer induced by cigarette smoking has increased over time. Long noncoding (lnc) RNAs, regulatory factors that play a role in human diseases, are commonly dysregulated in lung cancer. Cigarette smoking is closely related to changes in lncRNA expression, which can affect lung cancer. Herein, we assess the mechanism of lung cancer initiation induced by smoking. To calculate the impact of smoking on the survival of patients with lung cancer, we extracted data from The Cancer Genome Atlas and Gene Expression Omnibus databases and identified the differentially expressed genes in the lung cancer tissue compared to the normal lung tissue. Genes positively and negatively associated with smoking were identified. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and Cytoscape analyses were performed to determine the function of the genes and the effects of smoking on the immune microenvironment. lncRNAs corresponding to smoking-associated genes were identified, and a smoking-related lncRNA model was constructed using univariate and multivariate Cox analyses. This model was used to assess the survival of and potential risk in patients who smoked. During screening, 562 differentially expressed genes were identified, and we elucidated that smoking affected the survival of patients 4.5 years after the diagnosis of lung cancer. Furthermore, genes negatively associated with smoking were closely associated with immunity. Twelve immune cell types were also found to infiltrate differentially in smokers and nonsmokers. Thus, the smoking-associated lncRNA model is a good predictor of survival and risk in smokers and may be used as an independent prognostic factor for lung cancer.
Collapse
|
25
|
Eriksson Ström J, Kebede Merid S, Pourazar J, Blomberg A, Lindberg A, Ringh MV, Hagemann-Jensen M, Ekström TJ, Behndig AF, Melén E. COPD is Associated with Epigenome-wide Differential Methylation in BAL Lung Cells. Am J Respir Cell Mol Biol 2022; 66:638-647. [PMID: 35286818 PMCID: PMC9163645 DOI: 10.1165/rcmb.2021-0403oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
DNA methylation patterns in chronic pulmonary obstructive disease (COPD) might offer new insights into disease pathogenesis. To assess methylation profiles in the main COPD target organ, we performed an epigenome-wide association study on bronchoalveolar lavage (BAL) cells. Bronchoscopies were performed in 18 COPD subjects and 15 controls (ex- and current smokers). DNA methylation was measured with Illumina MethylationEPIC BeadChip covering >850,000 CpGs. Differentially methylated positions (DMPs) were examined for 1) enrichment in pathways and functional gene relationships using Kyoto Encyclopedia of Genes and Genomes and Gene Ontology; 2) accelerated aging using Horvath's epigenetic clock; 3) correlation with gene expression; and 4) co-localization with genetic variation. We found 1,155 Bonferroni significant (P < 6.74 × 10-8) DMPs associated with COPD, many with large effect sizes. Functional analysis identified biologically plausible pathways and gene relationships, including enrichment for transcription factor activity. Strong correlation was found between COPD and chronological age, but not with accelerated epigenetic aging. For 79 unique DMPs, DNA methylation correlated significantly with gene expression in BAL cells. Thirty-nine percent of DMPs were co-localized with COPD-associated SNPs. To the best of our knowledge, this is the first EWAS of COPD on BAL cells, and our analyses revealed many differential methylation sites. Integration with mRNA data showed a strong functional readout for relevant genes, identifying sites where DNA methylation might directly impact expression. Almost half of DMPs were co-located with SNPs identified in previous GWAS of COPD, suggesting joint genetic and epigenetic pathways related to disease.
Collapse
Affiliation(s)
- Jonas Eriksson Ström
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, Umeå, Sweden;
| | - Simon Kebede Merid
- Karolinska Institutet, 27106, Institute of Environmental Medicine, Stockholm, Sweden
| | - Jamshid Pourazar
- Umeå Universitet Medicinska fakulteten, 59588, Dept. of Public Health and Clinical Medicine, Umeå, Sweden
| | - Anders Blomberg
- Umea University, 8075, Dept. of Public Health and Clinical Medicine, Umea, Sweden
| | - Anne Lindberg
- Umeå Universitet, 8075, Department of Public Health and Clinical Medicine, Section of Medicine, Umea, Sweden
| | - Mikael V Ringh
- Karolinska Institutet, 27106, Department of Clinical Neuroscience and Center for Molecular Medicine, Stockholm, Sweden
| | | | - Tomas J Ekström
- Karolinska Institutet, 27106, Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Stockholm, Sweden
| | - Annelie F Behndig
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, Umeå, Sweden
| | - Erik Melén
- Karolinska Institutet Department of Clinical Science and Education Sodersjukhuset, 411435, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
26
|
Cardenas A, Ecker S, Fadadu RP, Huen K, Orozco A, McEwen LM, Engelbrecht HR, Gladish N, Kobor MS, Rosero-Bixby L, Dow WH, Rehkopf DH. Epigenome-wide association study and epigenetic age acceleration associated with cigarette smoking among Costa Rican adults. Sci Rep 2022; 12:4277. [PMID: 35277542 PMCID: PMC8917214 DOI: 10.1038/s41598-022-08160-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Smoking-associated DNA methylation (DNAm) signatures are reproducible among studies of mostly European descent, with mixed evidence if smoking accelerates epigenetic aging and its relationship to longevity. We evaluated smoking-associated DNAm signatures in the Costa Rican Study on Longevity and Healthy Aging (CRELES), including participants from the high longevity region of Nicoya. We measured genome-wide DNAm in leukocytes, tested Epigenetic Age Acceleration (EAA) from five clocks and estimates of telomere length (DNAmTL), and examined effect modification by the high longevity region. 489 participants had a mean (SD) age of 79.4 (10.8) years, and 18% were from Nicoya. Overall, 7.6% reported currently smoking, 35% were former smokers, and 57.4% never smoked. 46 CpGs and five regions (e.g. AHRR, SCARNA6/SNORD39, SNORA20, and F2RL3) were differentially methylated for current smokers. Former smokers had increased Horvath's EAA (1.69-years; 95% CI 0.72, 2.67), Hannum's EAA (0.77-years; 95% CI 0.01, 1.52), GrimAge (2.34-years; 95% CI1.66, 3.02), extrinsic EAA (1.27-years; 95% CI 0.34, 2.21), intrinsic EAA (1.03-years; 95% CI 0.12, 1.94) and shorter DNAmTL (- 0.04-kb; 95% CI - 0.08, - 0.01) relative to non-smokers. There was no evidence of effect modification among residents of Nicoya. Our findings recapitulate previously reported and novel smoking-associated DNAm changes in a Latino cohort.
Collapse
Affiliation(s)
- Andres Cardenas
- Division of Environmental Health Sciences, School of Public Health and Center for Computational Biology, University of California, Berkeley, 2121 Berkeley Way, #5121, Berkeley, CA, 94720, USA.
| | - Simone Ecker
- UCL Cancer Institute, University College London, London, UK
| | - Raj P Fadadu
- School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Karen Huen
- Division of Environmental Health Sciences, School of Public Health and Center for Computational Biology, University of California, Berkeley, 2121 Berkeley Way, #5121, Berkeley, CA, 94720, USA
| | - Allan Orozco
- School of Health Technology, Faculty of Medicine, University of Costa Rica (UCR), San José, San Pedro, Costa Rica
| | - Lisa M McEwen
- Faculty of Human and Social Development, School of Health Information Science, University of Victoria, Victoria, BC, Canada
| | - Hannah-Ruth Engelbrecht
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, and BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Nicole Gladish
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, and BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Michael S Kobor
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, and BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Luis Rosero-Bixby
- Centro Centroamericano de Población (CCP), Universidad de Costa Rica, San José, Costa Rica
| | - William H Dow
- Division of Health Policy and Management, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - David H Rehkopf
- Department of Epidemiology and Population Health and Department of Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| |
Collapse
|
27
|
Jacobsen KK, Schnohr P, Jensen GB, Bojesen SE. AHRR (cg5575921) methylation safely improves specificity of lung cancer screening eligibility criteria: A cohort study. Cancer Epidemiol Biomarkers Prev 2022; 31:758-765. [DOI: 10.1158/1055-9965.epi-21-1059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/19/2021] [Accepted: 01/04/2022] [Indexed: 11/16/2022] Open
|
28
|
Andersen A, Reimer R, Dawes K, Becker A, Hutchens N, Miller S, Dogan M, Hundley B, A Mills J, D Long J, Philibert R. DNA methylation differentiates smoking from vaping and non-combustible tobacco use. Epigenetics 2022; 17:178-190. [PMID: 33588690 PMCID: PMC8865289 DOI: 10.1080/15592294.2021.1890875] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/08/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Increasing use of non-combusted forms of nicotine such as e-cigarettes poses important public health questions regarding their specific risks relative to combusted tobacco products such as cigarettes. To fully delineate these risks, improved biomarkers that can distinguish between these forms of nicotine use are needed. Prior work has suggested that methylation status at cg05575921 may serve as a specific biomarker of combusted tobacco smoke exposure. We hypothesized combining this epigenetic biomarker with conventional metabolite assays could classify the type of nicotine product consumption. Therefore, we determined DNA methylation and serum cotinine values in samples from 112 smokers, 35 e-cigarette users, 19 smokeless tobacco users, and 269 controls, and performed mass spectroscopy analyses of urine samples from all nicotine users and 22 verified controls to determine urinary levels of putatively nicotine product-specific substances; propylene glycol, 2-cyanoethylmercapturic acid (CEMA), and anabasine. 1) Cigarette smoking was associated with a dose dependent demethylation of cg05575921 and increased urinary CEMA and anabasine levels, 2) e-cigarette use did not demethylate cg05575921, 3) smokeless tobacco use also did not demethylate cg05575921 but was positively associated with anabasine levels 4) CEMA and cg05575921 levels were highly correlated and 5) propylene glycol levels did not reliably distinguish use groups. Cg05575921 assessments distinguish exposure to tobacco smoke from smokeless sources of nicotine including e-cigarettes and smokeless tobacco, neither of which are associated with cg05575921 demethylation. A combination of methylomic and metabolite profiling may allow for accurate classification use status of a variety of nicotine containing products.
Collapse
Affiliation(s)
- Allan Andersen
- Department of Psychiatry, University of Iowa, Iowa City, USA
| | - Rachel Reimer
- College of Public Health, Des Moines University, Des Moines, USA
| | - Kelsey Dawes
- Department of Psychiatry, University of Iowa, Iowa City, USA
| | - Ashley Becker
- Department of Psychiatry, University of Iowa, Iowa City, USA
| | | | | | - Meesha Dogan
- Department of Psychiatry, University of Iowa, Iowa City, USA
- Behavioral Diagnostics LLC, Coralville, USA
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, United States
| | - Brandon Hundley
- Department of Psychiatry, University of Iowa, Iowa City, USA
| | - James A Mills
- Department of Psychiatry, University of Iowa, Iowa City, USA
| | - Jeffrey D Long
- Department of Psychiatry, University of Iowa, Iowa City, USA
- Department of Biostatistics, University of Iowa, Iowa City, USA
| | - Robert Philibert
- Department of Psychiatry, University of Iowa, Iowa City, USA
- Behavioral Diagnostics LLC, Coralville, USA
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, United States
| |
Collapse
|
29
|
Madhu NR, Sarkar B, Slama P, Jha NK, Ghorai SK, Jana SK, Govindasamy K, Massanyi P, Lukac N, Kumar D, Kalita JC, Kesari KK, Roychoudhury S. Effect of Environmental Stressors, Xenobiotics, and Oxidative Stress on Male Reproductive and Sexual Health. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1391:33-58. [PMID: 36472815 DOI: 10.1007/978-3-031-12966-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This article examines the environmental factor-induced oxidative stress (OS) and their effects on male reproductive and sexual health. There are several factors that induce OS, i.e. radition, metal contamination, xenobiotic compounds, and cigarette smoke and lead to cause toxicity in the cells through metabolic or bioenergetic processes. These environmental factors may produce free radicals and enhance the reactive oxygen species (ROS). Free radicals are molecules that include oxygen and disbalance the amount of electrons that can create major chemical chains in the body and cause oxidation. Oxidative damage to cells may impair male fertility and lead to abnormal embryonic development. Moreover, it does not only cause a vast number of health issues such as ageing, cancer, atherosclerosis, insulin resistance, diabetes mellitus, cardiovascular diseases, ischemia-reperfusion injury, and neurodegenerative disorders but also decreases the motility of spermatozoa while increasing sperm DNA damage, impairing sperm mitochondrial membrane lipids and protein kinases. This chapter mainly focuses on the environmental stressors with further discussion on the mechanisms causing congenital impairments due to poor sexual health and transmitting altered signal transduction pathways in male gonadal tissues.
Collapse
Affiliation(s)
- Nithar Ranjan Madhu
- Department of Zoology, Acharya Prafulla Chandra College, New Barrackpore, Kolkata, West Bengal, India
| | - Bhanumati Sarkar
- Department of Botany, Acharya Prafulla Chandra College, New Barrackpore, Kolkata, West Bengal, India
| | - Petr Slama
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, India
| | | | - Sandip Kumar Jana
- Department of Zoology, Bajkul Milani Mahavidyalaya, Purba Medinipur, West Bengal, India
| | - Kadirvel Govindasamy
- Animal Production Division, ICAR Research Complex for NEH Region, Indian Council of Agricultural Research, Umiam, Meghalaya, India
| | - Peter Massanyi
- Department of Animal Physiology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovak Republic
| | - Norbert Lukac
- Department of Animal Physiology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovak Republic
| | - Dhruv Kumar
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand, India
| | - Jogen C Kalita
- Department of Zoology, Gauhati University, Guwahati, India
| | | | | |
Collapse
|
30
|
Mao Y, Huang P, Wang Y, Wang M, Li MD, Yang Z. Genome-wide methylation and expression analyses reveal the epigenetic landscape of immune-related diseases for tobacco smoking. Clin Epigenetics 2021; 13:215. [PMID: 34886889 PMCID: PMC8662854 DOI: 10.1186/s13148-021-01208-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Smoking is a major causal risk factor for lung cancer, chronic obstructive pulmonary disease (COPD), cardiovascular disease (CVD), and is the main preventable cause of deaths in the world. The components of cigarette smoke are involved in immune and inflammatory processes, which may increase the prevalence of cigarette smoke-related diseases. However, the underlying molecular mechanisms linking smoking and diseases have not been well explored. This study was aimed to depict a global map of DNA methylation and gene expression changes induced by tobacco smoking and to explore the molecular mechanisms between smoking and human diseases through whole-genome bisulfite sequencing (WGBS) and RNA-sequencing (RNA-seq). RESULTS We performed WGBS on 72 samples (36 smokers and 36 nonsmokers) and RNA-seq on 75 samples (38 smokers and 37 nonsmokers), and cytokine immunoassay on plasma from 22 males (9 smokers and 13 nonsmokers) who were recruited from the city of Jincheng in China. By comparing the data of the two groups, we discovered a genome-wide methylation landscape of differentially methylated regions (DMRs) associated with smoking. Functional enrichment analyses revealed that both smoking-related hyper-DMR genes (DMGs) and hypo-DMGs were related to synapse-related pathways, whereas the hypo-DMGs were specifically related to cancer and addiction. The differentially expressed genes (DEGs) revealed by RNA-seq analysis were significantly enriched in the "immunosuppression" pathway. Correlation analysis of DMRs with their corresponding gene expression showed that genes affected by tobacco smoking were mostly related to immune system diseases. Finally, by comparing cytokine concentrations between smokers and nonsmokers, we found that vascular endothelial growth factor (VEGF) was significantly upregulated in smokers. CONCLUSIONS In sum, we found that smoking-induced DMRs have different distribution patterns in hypermethylated and hypomethylated areas between smokers and nonsmokers. We further identified and verified smoking-related DMGs and DEGs through multi-omics integration analysis of DNA methylome and transcriptome data. These findings provide us a comprehensive genomic map of the molecular changes induced by smoking which would enhance our understanding of the harms of smoking and its relationship with diseases.
Collapse
Affiliation(s)
- Ying Mao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Maiqiu Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming D Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Research Center for Air Pollution and Health, Zhejiang University, Hangzhou, China.
| | - Zhongli Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
31
|
Head and Neck Cancers Are Not Alike When Tarred with the Same Brush: An Epigenetic Perspective from the Cancerization Field to Prognosis. Cancers (Basel) 2021; 13:cancers13225630. [PMID: 34830785 PMCID: PMC8616074 DOI: 10.3390/cancers13225630] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Squamous cell carcinomas affect different head and neck subsites and, although these tumors arise from the same epithelial lining and share risk factors, they differ in terms of clinical behavior and molecular carcinogenesis mechanisms. Differences between HPV-negative and HPV-positive tumors are those most frequently explored, but further data suggest that the molecular heterogeneity observed among head and neck subsites may go beyond HPV infection. In this review, we explore how alterations of DNA methylation and microRNA expression contribute to head and neck squamous cell carcinoma (HNSCC) development and progression. The association of these epigenetic alterations with risk factor exposure, early carcinogenesis steps, transformation risk, and prognosis are described. Finally, we discuss the potential application of the use of epigenetic biomarkers in HNSCC. Abstract Head and neck squamous cell carcinomas (HNSCC) are among the ten most frequent types of cancer worldwide and, despite all efforts, are still diagnosed at late stages and show poor overall survival. Furthermore, HNSCC patients often experience relapses and the development of second primary tumors, as a consequence of the field cancerization process. Therefore, a better comprehension of the molecular mechanisms involved in HNSCC development and progression may enable diagnosis anticipation and provide valuable tools for prediction of prognosis and response to therapy. However, the different biological behavior of these tumors depending on the affected anatomical site and risk factor exposure, as well as the high genetic heterogeneity observed in HNSCC are major obstacles in this pursue. In this context, epigenetic alterations have been shown to be common in HNSCC, to discriminate the tumor anatomical subsites, to be responsive to risk factor exposure, and show promising results in biomarker development. Based on this, this review brings together the current knowledge on alterations of DNA methylation and microRNA expression in HNSCC natural history, focusing on how they contribute to each step of the process and on their applicability as biomarkers of exposure, HNSCC development, progression, and response to therapy.
Collapse
|
32
|
Richmond RC, Sillero-Rejon C, Khouja JN, Prince C, Board A, Sharp G, Suderman M, Relton CL, Munafò M, Gage SH. Investigating the DNA methylation profile of e-cigarette use. Clin Epigenetics 2021; 13:183. [PMID: 34583751 PMCID: PMC8479883 DOI: 10.1186/s13148-021-01174-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 09/18/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Little evidence exists on the health effects of e-cigarette use. DNA methylation may serve as a biomarker for exposure and could be predictive of future health risk. We aimed to investigate the DNA methylation profile of e-cigarette use. RESULTS Among 117 smokers, 117 non-smokers and 116 non-smoking vapers, we evaluated associations between e-cigarette use and epigenome-wide methylation from saliva. DNA methylation at 7 cytosine-phosphate-guanine sites (CpGs) was associated with e-cigarette use at p < 1 × 10-5 and none at p < 5.91 × 10-8. 13 CpGs were associated with smoking at p < 1 × 10-5 and one at p < 5.91 × 10-8. CpGs associated with e-cigarette use were largely distinct from those associated with smoking. There was strong enrichment of known smoking-related CpGs in the smokers but not the vapers. We also tested associations between e-cigarette use and methylation scores known to predict smoking and biological ageing. Methylation scores for smoking and biological ageing were similar between vapers and non-smokers. Higher levels of all smoking scores and a biological ageing score (GrimAge) were observed in smokers. A methylation score for e-cigarette use showed poor prediction internally (AUC 0.55, 0.41-0.69) and externally (AUC 0.57, 0.36-0.74) compared with a smoking score (AUCs 0.80) and was less able to discriminate lung squamous cell carcinoma from adjacent normal tissue (AUC 0.64, 0.52-0.76 versus AUC 0.73, 0.61-0.85). CONCLUSIONS The DNA methylation profile for e-cigarette use is largely distinct from that of cigarette smoking, did not replicate in independent samples, and was unable to discriminate lung cancer from normal tissue. The extent to which methylation related to long-term e-cigarette use translates into chronic effects requires further investigation.
Collapse
Affiliation(s)
- Rebecca C Richmond
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.
| | - Carlos Sillero-Rejon
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- National Institute for Health Research Applied Research Collaboration West (NIHR ARC West), University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Jasmine N Khouja
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- School of Psychological Science, University of Bristol, Bristol, UK
| | - Claire Prince
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Alexander Board
- Department of Experimental Psychology, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Gemma Sharp
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Matthew Suderman
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Marcus Munafò
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- School of Psychological Science, University of Bristol, Bristol, UK
| | - Suzanne H Gage
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Department of Psychological Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
33
|
Buck JM, Yu L, Knopik VS, Stitzel JA. DNA methylome perturbations: an epigenetic basis for the emergingly heritable neurodevelopmental abnormalities associated with maternal smoking and maternal nicotine exposure†. Biol Reprod 2021; 105:644-666. [PMID: 34270696 PMCID: PMC8444709 DOI: 10.1093/biolre/ioab138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/29/2021] [Accepted: 07/14/2021] [Indexed: 11/13/2022] Open
Abstract
Maternal smoking during pregnancy is associated with an ensemble of neurodevelopmental consequences in children and therefore constitutes a pressing public health concern. Adding to this burden, contemporary epidemiological and especially animal model research suggests that grandmaternal smoking is similarly associated with neurodevelopmental abnormalities in grandchildren, indicative of intergenerational transmission of the neurodevelopmental impacts of maternal smoking. Probing the mechanistic bases of neurodevelopmental anomalies in the children of maternal smokers and the intergenerational transmission thereof, emerging research intimates that epigenetic changes, namely DNA methylome perturbations, are key factors. Altogether, these findings warrant future research to fully elucidate the etiology of neurodevelopmental impairments in the children and grandchildren of maternal smokers and underscore the clear potential thereof to benefit public health by informing the development and implementation of preventative measures, prophylactics, and treatments. To this end, the present review aims to encapsulate the burgeoning evidence linking maternal smoking to intergenerational epigenetic inheritance of neurodevelopmental abnormalities, to identify the strengths and weaknesses thereof, and to highlight areas of emphasis for future human and animal model research therein.
Collapse
Affiliation(s)
- Jordan M Buck
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO, USA
| | - Li Yu
- Department of Human Development and Family Studies, Purdue University, West Lafayette, IN, USA
| | - Valerie S Knopik
- Department of Human Development and Family Studies, Purdue University, West Lafayette, IN, USA
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Integrative Physiology, University of Colorado, Boulder, Boulder, CO, USA
| |
Collapse
|
34
|
Suhre K, Zaghlool S. Connecting the epigenome, metabolome and proteome for a deeper understanding of disease. J Intern Med 2021; 290:527-548. [PMID: 33904619 DOI: 10.1111/joim.13306] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 12/26/2022]
Abstract
Epigenome-wide association studies (EWAS) identify genes that are dysregulated by the studied clinical endpoints, thereby indicating potential new diagnostic biomarkers, drug targets and therapy options. Combining EWAS with deep molecular phenotyping, such as approaches enabled by metabolomics and proteomics, allows further probing of the underlying disease-associated pathways. For instance, methylation of the TXNIP gene is associated robustly with prevalent type 2 diabetes and further with metabolites that are short-term markers of glycaemic control. These associations reflect TXNIP's function as a glucose uptake regulator by interaction with the major glucose transporter GLUT1 and suggest that TXNIP methylation can be used as a read-out for the organism's exposure to glucose stress. Another case is the association between DNA methylation of the AHRR and F2RL3 genes with smoking and a protein that is involved in the reprogramming of the bronchial epithelium. These examples show that associations between DNA methylation and intermediate molecular traits can open new windows into how the body copes with physiological challenges. This knowledge, if carefully interpreted, may indicate novel therapy options and, together with monitoring of the methylation state of specific methylation sites, may in the future allow the early diagnosis of impending disease. It is essential for medical practitioners to recognize the potential that this field holds in translating basic research findings to clinical practice. In this review, we present recent advances in the field of EWAS with metabolomics and proteomics and discuss both the potential and the challenges of translating epigenetic associations, with deep molecular phenotypes, to biomedical applications.
Collapse
Affiliation(s)
- K Suhre
- From the, Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar.,Department of Biophysics and Physiology, Weill Cornell Medicine, New York, USA
| | - S Zaghlool
- From the, Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar.,Department of Biophysics and Physiology, Weill Cornell Medicine, New York, USA
| |
Collapse
|
35
|
Huo X, Jin S, Wang Y, Ma L. DNA methylation in chronic obstructive pulmonary disease. Epigenomics 2021; 13:1145-1155. [PMID: 34142873 DOI: 10.2217/epi-2021-0111] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD), a complex disease with polygenetic tendency, is one of the most important health problems in the world. Recently, in the study of the pathogenesis of the COPD, epigenetic changes caused by environmental factors, such as DNA methylation, started to attract more attention than genetic factors. In this review, we discuss the main features of DNA methylation, such as DNA methyltransferases and the methylation sites that modulate the DNA methylation level, and their roles in COPD progression. Finally, to promote new ideas for the prevention and treatment of COPD, we focus on the potential of DNA methylation as a COPD therapeutic target.
Collapse
Affiliation(s)
- XinXin Huo
- School of Public Health, Lanzhou University, Lanzhou, China
| | - SiHui Jin
- School of Public Health, Lanzhou University, Lanzhou, China
| | - YiGe Wang
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Li Ma
- School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
36
|
Chung YK, Kim JJ, Hong MA, Hwang KR, Chae SJ, Yoon SH, Choi YM. Association Between Polycystic Ovary Syndrome and the Polymorphisms of Aryl Hydrocarbon Receptor Repressor, Glutathione-S-transferase T1, and Glutathione-S-transferase M1 Genes. Gynecol Endocrinol 2021; 37:558-561. [PMID: 33124502 DOI: 10.1080/09513590.2020.1832066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVE To investigate the association between the aryl hydrocarbon receptor repressor (AhRR) C/G polymorphisms and glutathione-S-transferase M1 (GSTM1) and GSTT1 null mutation and the risk of polycystic ovary syndrome (PCOS) in Korean women. METHODS This was a case-control study of 478 women with PCOS and 376 aged-matched healthy controls. Genotyping of the AhRR C/G polymorphism and GSTM1 and GSTT1 were performed using real-time PCR analysis and multiplex PCR, respectively. RESULTS The genotype distribution of the AhRR C/G polymorphisms and GSTM1/GSTT1 null mutations did not differ between women with PCOS and controls. Using the wild-type combined AhRR CC and GSTT1 present genotype as a reference, the odds that a woman had PCOS were 1.54 (95% CIs 1.04-2.29) times higher if she had a combined AhRR CG or GG and GSTT1 null genotype. The odds that a woman had PCOS was 1.48 (95% CIs 1.08-2.04) times higher if she had a combined GSTM1/GSTT1 null genotype compared with the wild-type combined GSTM1/GSTT1 present genotype. However, there were no significant associations between the risk of PCOS and any combined AhRR and GSTM1. CONCLUSIONS Our data suggest that a combined AhRR CG or GG and GSTT1 null genotype or a combined GSTT1/GSTM1 null genotype might be associated with an increased risk of PCOS.
Collapse
Affiliation(s)
- Youn Kyung Chung
- Department of Obstetrics and Gynecology, National Cancer Center, Goyang, Korea
| | - Jin Ju Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
- Department of Obstetrics and Gynecology, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Korea
- The Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Min A Hong
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Kyu Ri Hwang
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
- Department of Obstetrics and Gynecology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Soo Jin Chae
- Department of Obstetrics and Gynecology, IORA Fertility Clinic, Suwon, Korea
| | - Sang Ho Yoon
- Department of Obstetrics and Gynecology, IORA Fertility Clinic, Suwon, Korea
| | - Young Min Choi
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
- The Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
37
|
Reddy KD, Lan A, Boudewijn IM, Rathnayake SNH, Koppelman GH, Oliver BG, van den Berge M, Faiz A. Current-Smoking alters Gene Expression and DNA Methylation in the Nasal Epithelium of Asthmatics. Am J Respir Cell Mol Biol 2021; 65:366-377. [PMID: 33989148 DOI: 10.1165/rcmb.2020-0553oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Current-smoking contributes to worsened asthma prognosis, more severe symptoms and limits the beneficial effects of corticosteroids. As the nasal epithelium can reflect smoking-induced changes in the lower airways, it is a relevant source to investigate changes in gene expression and DNA methylation. This study explores gene expression and DNA methylation changes in current and ex-smokers with asthma. Matched gene expression and epigenome-wide DNA methylation samples collected from nasal brushings of 55 patients enrolled in a clinical trial investigation of current and ex-smoker asthma patients were analysed. Differential gene expression and DNA methylation analyses were conducted comparing current- vs ex-smokers. Expression quantitative trait methylation (eQTM) analysis was completed to explore smoking relevant genes by CpG sites that differ between current and ex-smokers. To investigate the relevance of the smoking-associated DNA methylation changes for the lower airways, significant CpG sites were explored in bronchial biopsies from patients who had stopped smoking. 809 genes and 18,814 CpG sites were differentially associated with current-smoking in the nose. The cis-eQTM analysis uncovered 171 CpG sites whose methylation status associated with smoking-related gene expression, including AHRR, ALDH3A1, CYP1A1 and CYP1B1. Methylation status of CpG sites altered by current-smoking reversed with one-year smoking cessation. We confirm current-smoking alters epigenetic patterns and affects gene expression in the nasal epithelium of asthma patients, which is partially reversible in bronhcial biopsies after smoking cessation. We demonstrate the ability to discern molecular changes in the nasal epithelium, presenting this as a tool in future investigations into disease-relevant effects of tobacco smoke.
Collapse
Affiliation(s)
- Karosham D Reddy
- Woolcock Institute of Medical Research, 104349, Cell Biology, Glebe, New South Wales, Australia.,University of Technology Sydney, 1994, School of Life Sciences, Ultimo, New South Wales, Australia;
| | - Andy Lan
- University of Groningen, 3647, Department of Pulmonary Diseases, Groningen, Netherlands.,University of Groningen, 3647, GRIAC Research Institute, Groningen, Netherlands
| | - Ilse M Boudewijn
- University of Groningen, 3647, Department of Pulmonary Diseases, Groningen, Netherlands.,University of Groningen, 3647, GRIAC Research Insitute, Groningen, Netherlands
| | - Senani N H Rathnayake
- University of Technology Sydney, 1994, Respiratory Bioinformatics and Molecular Biology (RBMB) group,, Sydney, New South Wales, Australia
| | - Gerard H Koppelman
- University of Groningen, 3647, University Medical Center Groningen, Department of Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, Groningen, Netherlands.,University of Groningen, 3647, University Medical Center Groningen, GRIAC research institute, Groningen, Netherlands
| | - Brian G Oliver
- Woolcock Institute of Medical Research, 104349, Glebe, New South Wales, Australia.,University of Technology Sydney, 1994, School of Medical and Molecular Biosciences, Sydney, New South Wales, Australia
| | - Maarten van den Berge
- University of Groningen, 3647, University Medical Center, Department of Pulmonary Diseases, Groningen, Netherlands.,University Medical Center Göttingen, 84922, Groningen Research Institute for Asthma and COPD (GRIAC), Gottingen, Germany
| | - Alen Faiz
- University of Technology Sydney, 1994, Respiratory Bioinformatics and Molecular Biology (RBMB), School of Life Sciences, Sydney, New South Wales, Australia
| | | |
Collapse
|
38
|
Fuemmeler BF, Dozmorov MG, Do EK, Zhang J(J, Grenier C, Huang Z, Maguire RL, Kollins SH, Hoyo C, Murphy SK. DNA Methylation in Babies Born to Nonsmoking Mothers Exposed to Secondhand Smoke during Pregnancy: An Epigenome-Wide Association Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:57010. [PMID: 34009014 PMCID: PMC8132610 DOI: 10.1289/ehp8099] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 02/09/2021] [Accepted: 04/19/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Maternal smoking during pregnancy is related to altered DNA methylation in infant umbilical cord blood. The extent to which low levels of smoke exposure among nonsmoking pregnant women relates to offspring DNA methylation is unknown. OBJECTIVE This study sought to evaluate relationships between maternal prenatal plasma cotinine levels and DNA methylation in umbilical cord blood in newborns using the Infinium HumanMethylation 450K BeadChip. METHODS Participants from the Newborn Epigenetics Study cohort who reported not smoking during pregnancy had verified low levels of cotinine from maternal prenatal plasma (0 ng / mL to < 4 ng / mL ), and offspring epigenetic data from umbilical cord blood were included in this study (n = 79 ). Multivariable linear regression models were fit to the data, controlling for cell proportions, age, race, education, and parity. Estimates represent changes in response to any 1 -ng / mL unit increase in exposure. RESULTS Multivariable linear regression models yielded 29,049 CpGs that were differentially methylated in relation to increases in cotinine at a 5% false discovery rate. Top CpGs were within or near genes involved in neuronal functioning (PRKG1, DLGAP2, BSG), carcinogenesis (FHIT, HSPC157) and inflammation (AGER). Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses suggest cotinine was related to methylation of gene pathways controlling neuronal signaling, metabolic regulation, cell signaling and regulation, and cancer. Further, enhancers associated with transcription start sites were enriched in altered CpGs. Using an independent sample from the same study population (n = 115 ), bisulfite pyrosequencing was performed with infant cord blood DNA for two genes within our top 20 hits (AGER and PRKG1). Results from pyrosequencing replicated epigenome results for PRKG1 (cg17079497, estimate = - 1.09 , standard error ( SE ) = 0.45 , p = 0.018 ) but not for AGER (cg09199225; estimate = - 0.16 , SE = 0.21 , p = 0.44 ). DISCUSSION Secondhand smoke exposure among nonsmoking women may alter DNA methylation in regions involved in development, carcinogenesis, and neuronal functioning. These novel findings suggest that even low levels of smoke exposure during pregnancy may be sufficient to alter DNA methylation in distinct sites of mixed umbilical cord blood leukocytes in pathways that are known to be altered in cord blood from pregnant active smokers. https://doi.org/10.1289/EHP8099.
Collapse
Affiliation(s)
- Bernard F. Fuemmeler
- Department of Health Behavior and Policy, Virginia Commonwealth University, Richmond, Virginia, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Mikhail G. Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Pathology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Elizabeth K. Do
- Department of Health Behavior and Policy, Virginia Commonwealth University, Richmond, Virginia, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Junfeng (Jim) Zhang
- Nicholas School of the Environment and Duke Global Health Institute, Duke University, Durham, North Carolina, USA
| | - Carole Grenier
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina, USA
| | - Zhiqing Huang
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina, USA
| | - Rachel L. Maguire
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Biological Sciences, Center for Human Health and the Environment North Carolina State University, Raleigh, North Carolina, USA
| | - Scott H. Kollins
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, USA
| | - Cathrine Hoyo
- Department of Biological Sciences, Center for Human Health and the Environment North Carolina State University, Raleigh, North Carolina, USA
| | - Susan K. Murphy
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
39
|
Jung Y, Frisvold D, Dogan T, Dogan M, Philibert R. Cost-utility analysis of an integrated genetic/epigenetic test for assessing risk for coronary heart disease. Epigenomics 2021; 13:531-547. [PMID: 33625255 DOI: 10.2217/epi-2021-0021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Aim: New epigenetically based methods for assessing risk for coronary heart disease may be more sensitive but are generally more costly than current methods. To understand their potential impact on healthcare spending, we conducted a cost-utility analysis. Methods: We compared costs using the new Epi + Gen CHD™ test with those of existing tests using a cohort Markov simulation model. Results: We found that use of the new test was associated with both better survival and highly competitive negative incremental cost-effectiveness ratios ranging from -$42,000 to -$8000 per quality-adjusted life year for models with and without a secondary test. Conclusion: The new integrated genetic/epigenetic test will save money and lives under most real-world scenarios. Similar advantages may be seen for other epigenetic tests.
Collapse
Affiliation(s)
- Younsoo Jung
- Cardio Diagnostics Inc., Coralville, IA 52241, USA
| | - David Frisvold
- Department of Economics, University of Iowa, Iowa City, IA 52242, USA
| | - Timur Dogan
- Cardio Diagnostics Inc., Coralville, IA 52241, USA
| | | | - Rob Philibert
- Cardio Diagnostics Inc., Coralville, IA 52241, USA.,Department of Psychiatry, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
40
|
Philibert R, Mills JA, Long JD, Salisbury SE, Comellas A, Gerke A, Dawes K, Vander Weg M, Hoffman EA. The Reversion of cg05575921 Methylation in Smoking Cessation: A Potential Tool for Incentivizing Healthy Aging. Genes (Basel) 2020; 11:E1415. [PMID: 33260961 PMCID: PMC7760261 DOI: 10.3390/genes11121415] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
Smoking is the largest preventable cause of mortality and the largest environmental driver of epigenetic aging. Contingency management-based strategies can be used to treat smoking but require objective methods of verifying quitting status. Prior studies have suggested that cg05575921 methylation reverts as a function of smoking cessation, but that it can be used to verify the success of smoking cessation has not been unequivocally demonstrated. To test whether methylation can be used to verify cessation, we determined monthly cg05575921 levels in a group of 67 self-reported smokers undergoing biochemically monitored contingency management-based smoking cessation therapy, as part of a lung imaging protocol. A total of 20 subjects in this protocol completed three months of cotinine verified smoking cessation. In these 20 quitters, the reversion of cg05575921 methylation was dependent on their initial smoking intensity, with methylation levels in the heaviest smokers reverting to an average of 0.12% per day over the 3-month treatment period. In addition, we found suggestive evidence that some individuals may have embellished their smoking history to gain entry to the study. Given the prominent effect of smoking on longevity, we conclude that DNA methylation may be a useful tool for guiding and incentivizing contingency management-based approaches for smoking cessation.
Collapse
Affiliation(s)
- Robert Philibert
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242, USA; (J.A.M.); (J.D.L.); (K.D.)
- Behavioral Diagnostics LLC, Coralville, IA 52241, USA
| | - James A. Mills
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242, USA; (J.A.M.); (J.D.L.); (K.D.)
| | - Jeffrey D. Long
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242, USA; (J.A.M.); (J.D.L.); (K.D.)
- Department of Biostatistics, University of Iowa, Iowa City, IA 52242, USA
| | - Sue Ellen Salisbury
- Department of Radiology, University of Iowa, Iowa City, IA 52242, USA; (S.E.S.); (E.A.H.)
| | - Alejandro Comellas
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA; (A.C.); (A.G.); (M.V.W.)
| | - Alicia Gerke
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA; (A.C.); (A.G.); (M.V.W.)
| | - Kelsey Dawes
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242, USA; (J.A.M.); (J.D.L.); (K.D.)
- Molecular Medicine Program, University of Iowa, Iowa City, IA 52242, USA
| | - Mark Vander Weg
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA; (A.C.); (A.G.); (M.V.W.)
- Center for Access & Delivery Research and Evaluation, Iowa City VA Health Care System, Iowa City, IA 52242, USA
| | - Eric A. Hoffman
- Department of Radiology, University of Iowa, Iowa City, IA 52242, USA; (S.E.S.); (E.A.H.)
| |
Collapse
|
41
|
Pittman GS, Wang X, Campbell MR, Coulter SJ, Olson JR, Pavuk M, Birnbaum LS, Bell DA. Dioxin-like compound exposures and DNA methylation in the Anniston Community Health Survey Phase II. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 742:140424. [PMID: 32629249 PMCID: PMC7574543 DOI: 10.1016/j.scitotenv.2020.140424] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/30/2020] [Accepted: 06/20/2020] [Indexed: 05/26/2023]
Abstract
The Anniston Community Health Survey (ACHS-I) was initially conducted from 2005 to 2007 to assess polychlorinated biphenyl (PCB) exposures in Anniston, Alabama residents. In 2014, a follow-up study (ACHS-II) was conducted to measure the same PCBs as in ACHS-I and additional compounds e.g., polychlorinated dibenzo-p-dioxins (PCDDs), polychlorinated dibenzofurans (PCDFs), and dioxin-like non-ortho (cPCBs) substituted PCBs. In this epigenome-wide association study (EWAS), we examined the associations between PCDD, PCDF, and PCB exposures and DNA methylation. Whole blood DNA methylation was measured using Illumina EPIC arrays (n=292). We modeled lipid-adjusted toxic equivalencies (TEQs) for: ΣDioxins (sum of 28 PCDDs, PCDFs, cPCBs, and mPCBs), PCDDs, PCDFs, cPCBs, and mPCBs using robust multivariable linear regression adjusting for age, race, sex, smoking, bisulfite conversion batch, and estimated percentages of six blood cell types. Among all exposures we identified 10 genome-wide (Bonferroni p≤6.74E-08) and 116 FDR (p≤5.00E-02) significant associations representing 10 and 113 unique CpGs, respectively. Of the 10 genome-wide associations, seven (70%) occurred in the PCDDs and four (40%) of these associations had an absolute differential methylation ≥1.00%, based on the methylation difference between the highest and lowest exposure quartiles. Most of the associations (six, 60%) represented hypomethylation changes. Of the 10 unique CpGs, eight (80%) were in genes shown to be associated with dioxins and/or PCBs based on data from the 2019 Comparative Toxicogenomics Database. In this study, we have identified a set of CpGs in blood DNA that may be particularly susceptible to dioxin, furan, and dioxin-like PCB exposures.
Collapse
Affiliation(s)
- Gary S Pittman
- National Institute of Environmental Health Sciences-National Institutes of Health, Research Triangle Park, NC 27709, United States of America
| | - Xuting Wang
- National Institute of Environmental Health Sciences-National Institutes of Health, Research Triangle Park, NC 27709, United States of America.
| | - Michelle R Campbell
- National Institute of Environmental Health Sciences-National Institutes of Health, Research Triangle Park, NC 27709, United States of America.
| | - Sherry J Coulter
- National Institute of Environmental Health Sciences-National Institutes of Health, Research Triangle Park, NC 27709, United States of America.
| | - James R Olson
- University at Buffalo, Buffalo, NY 14214, United States of America.
| | - Marian Pavuk
- Agency for Toxic Substances and Disease Registry, Centers for Disease Control and Prevention, Atlanta, GA 30341, United States of America.
| | - Linda S Birnbaum
- National Institute of Environmental Health Sciences-National Institutes of Health, Research Triangle Park, NC 27709, United States of America.
| | - Douglas A Bell
- National Institute of Environmental Health Sciences-National Institutes of Health, Research Triangle Park, NC 27709, United States of America.
| |
Collapse
|
42
|
Hulls PM, de Vocht F, Bao Y, Relton CL, Martin RM, Richmond RC. DNA methylation signature of passive smoke exposure is less pronounced than active smoking: The Understanding Society study. ENVIRONMENTAL RESEARCH 2020; 190:109971. [PMID: 32745538 PMCID: PMC7536273 DOI: 10.1016/j.envres.2020.109971] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/09/2020] [Accepted: 07/17/2020] [Indexed: 06/11/2023]
Abstract
INTRODUCTION The extent of the biological impact of passive smoke exposure is unclear. We sought to investigate the association between passive smoke exposure and DNA methylation, which could serve as a biomarker of health risk. MATERIALS AND METHODS We derived passive smoke exposure from self-reported questionnaire data among smoking and non-smoking partners of participants enrolled in the UK Household Longitudinal Study 'Understanding Society' (n=769). We performed an epigenome-wide association study (EWAS) of passive smoke exposure with DNA methylation in peripheral blood measured using the Illumina Infinium Methylation EPIC array. RESULTS No CpG sites surpassed the epigenome-wide significance threshold of p<5.97 × 10-8 in relation to partner smoking, compared with 10 CpG sites identified in relation to own smoking. However, 10 CpG sites surpassed a less stringent threshold of p<1 × 10-5 in a model of partner smoking adjusted for own smoking (model 1), 7 CpG sites in a model of partner smoking restricted to non-smokers (model 2) and 16 CpGs in a model restricted to regular smokers (model 3). In addition, there was evidence for an interaction between own smoking status and partners' smoking status on DNA methylation levels at the majority of CpG sites identified in models 2 and 3. There was a clear lack of enrichment for previously identified smoking signals in the EWAS of passive smoke exposure compared with the EWAS of own smoking. CONCLUSION The DNA methylation signature associated with passive smoke exposure is much less pronounced than that of own smoking, with no positive findings for 'expected' signals. It is unlikely that changes to DNA methylation serve as an important mechanism underlying the health risks of passive smoke exposure.
Collapse
Affiliation(s)
- Paige M Hulls
- Population Health Sciences, Bristol Medical School, University of Bristol, BS8 2BN, UK; MRC Integrative Epidemiology Unit at University of Bristol, BS8 2BN, UK.
| | - Frank de Vocht
- Population Health Sciences, Bristol Medical School, University of Bristol, BS8 2BN, UK; NIHR Applied Research Collaboration West (NIHR ARC West), Bristol, BS1 2NT, UK.
| | - Yanchun Bao
- Institute for Social and Economic Research, University of Essex, CO4 3SQ, UK.
| | - Caroline L Relton
- Population Health Sciences, Bristol Medical School, University of Bristol, BS8 2BN, UK; MRC Integrative Epidemiology Unit at University of Bristol, BS8 2BN, UK.
| | - Richard M Martin
- Population Health Sciences, Bristol Medical School, University of Bristol, BS8 2BN, UK; MRC Integrative Epidemiology Unit at University of Bristol, BS8 2BN, UK; NIHR Bristol Biomedical Research Centre at the University Hospitals Bristol NHS Foundation Trust and the University of Bristol, BS8 2BN, UK.
| | - Rebecca C Richmond
- Population Health Sciences, Bristol Medical School, University of Bristol, BS8 2BN, UK; MRC Integrative Epidemiology Unit at University of Bristol, BS8 2BN, UK.
| |
Collapse
|
43
|
Grieshober L, Graw S, Barnett MJ, Thornquist MD, Goodman GE, Chen C, Koestler DC, Marsit CJ, Doherty JA. AHRR methylation in heavy smokers: associations with smoking, lung cancer risk, and lung cancer mortality. BMC Cancer 2020; 20:905. [PMID: 32962699 PMCID: PMC7510160 DOI: 10.1186/s12885-020-07407-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/14/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A low level of methylation at cg05575921 in the aryl-hydrocarbon receptor repressor (AHRR) gene is robustly associated with smoking, and some studies have observed associations between cg05575921 methylation and increased lung cancer risk and mortality. To prospectively examine whether decreased methylation at cg05575921 may identify high risk subpopulations for lung cancer screening among heavy smokers, and mortality in cases, we evaluated associations between cg05575921 methylation and lung cancer risk and mortality, by histotype, in heavy smokers. METHODS The β-Carotene and Retinol Efficacy Trial (CARET) included enrollees ages 45-69 with ≥ 20 pack-year smoking histories and/or occupational asbestos exposure. A subset of CARET participants had cg05575921 methylation available from HumanMethylationEPIC assays of blood collected on average 4.3 years prior to lung cancer diagnosis in cases. Cg05575921 methylation β-values were treated continuously for a 10% methylation decrease and as quintiles, where quintile 1 (Q1, referent) represents high methylation and Q5, low methylation. We used conditional logistic regression models to examine lung cancer risk overall and by histotype in a nested case-control study including 316 lung cancer cases (diagnosed through 2005) and 316 lung cancer-free controls matched on age (±5 years), sex, race/ethnicity, enrollment year, current/former smoking, asbestos exposure, and follow-up time. Mortality analyses included 372 lung cancer cases diagnosed between 1985 and 2013 with available methylation data. We used Cox proportional hazards models to examine mortality overall and by histotype. RESULTS Decreased cg05575921 methylation was strongly associated with smoking, even in our population of heavy smokers. We did not observe associations between decreased pre-diagnosis cg05575921 methylation and increased lung cancer risk, overall or by histotype. We observed linear increasing trends for lung cancer-specific mortality across decreasing cg05575921 methylation quintiles for adenocarcinoma and small cell carcinoma (P-trends = 0.01 and 0.04, respectively). CONCLUSIONS In our study of heavy smokers, decreased cg05575921 methylation was strongly associated with smoking but not increased lung cancer risk. The observed association between cg05575921 methylation and increased mortality in adenocarcinoma and small cell histotypes requires further examination. Our results do not support using decreased cg05575921 methylation as a biomarker for lung cancer screening risk stratification.
Collapse
Affiliation(s)
- Laurie Grieshober
- Department of Population Health Sciences, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive, Room 4746, Salt Lake City, UT, 84112, USA.
| | - Stefan Graw
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Matt J Barnett
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Mark D Thornquist
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Gary E Goodman
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Chu Chen
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA.,Department of Otolaryngology: Head and Neck Surgery, School of Medicine, University of Washington, Seattle, WA, USA
| | - Devin C Koestler
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Carmen J Marsit
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jennifer A Doherty
- Department of Population Health Sciences, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive, Room 4746, Salt Lake City, UT, 84112, USA.,Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
44
|
You C, Wu S, Zheng SC, Zhu T, Jing H, Flagg K, Wang G, Jin L, Wang S, Teschendorff AE. A cell-type deconvolution meta-analysis of whole blood EWAS reveals lineage-specific smoking-associated DNA methylation changes. Nat Commun 2020; 11:4779. [PMID: 32963246 PMCID: PMC7508850 DOI: 10.1038/s41467-020-18618-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
Highly reproducible smoking-associated DNA methylation changes in whole blood have been reported by many Epigenome-Wide-Association Studies (EWAS). These epigenetic alterations could have important implications for understanding and predicting the risk of smoking-related diseases. To this end, it is important to establish if these DNA methylation changes happen in all blood cell subtypes or if they are cell-type specific. Here, we apply a cell-type deconvolution algorithm to identify cell-type specific DNA methylation signals in seven large EWAS. We find that most of the highly reproducible smoking-associated hypomethylation signatures are more prominent in the myeloid lineage. A meta-analysis further identifies a myeloid-specific smoking-associated hypermethylation signature enriched for DNase Hypersensitive Sites in acute myeloid leukemia. These results may guide the design of future smoking EWAS and have important implications for our understanding of how smoking affects immune-cell subtypes and how this may influence the risk of smoking related diseases. Smoking-associated DNA methylation changes in whole blood have been reported by many EWAS. Here, the authors use a cell-type deconvolution algorithm to identify cell-type specific DNA methylation signals in seven EWAS, identifying lineage-specific smoking-associated DNA methylation changes.
Collapse
Affiliation(s)
- Chenglong You
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Sijie Wu
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.,Human Phenome Institute, Fudan University, 825 Zhangheng Road, Shanghai, China.,State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Shijie C Zheng
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.,Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Tianyu Zhu
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Han Jing
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Ken Flagg
- Guangzhou Regenerative Medicine Guangdong Laboratory, Guangzhou, China
| | - Guangyu Wang
- Department of Computer Science and Technology, Tsinghua University, Beijing, China
| | - Li Jin
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.,Human Phenome Institute, Fudan University, 825 Zhangheng Road, Shanghai, China.,State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China. .,UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street, London, WC1E 6BT, UK.
| |
Collapse
|
45
|
AHRR hypomethylation as an epigenetic marker of smoking history predicts risk of myocardial infarction in former smokers. Atherosclerosis 2020; 312:8-15. [PMID: 32947224 DOI: 10.1016/j.atherosclerosis.2020.08.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/11/2020] [Accepted: 08/18/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS Smoking causes cardiovascular disease. AHRR hypomethylation at the cg05575921 site is associated with active and former smoking status at baseline, and cumulative amount of tobacco smoked. We tested the hypothesis that AHRR cg05575921 hypomethylation as an epigenetic marker of smoking history predicts the risk of myocardial infarction in former smokers. METHODS We included 10,510 individuals with methylation extent measurements and information on smoking status from the Copenhagen City Heart Study (CCHS), a prospective, cohort study of the general population carried out from 1991 to 2003. The endpoint myocardial infarction was retrieved from the national Danish Patient Registry and the national Danish Causes of Death Registry. RESULTS For individuals in the 1st (lowest) quartile of AHRR cg05575921 methylation (≤49% methylation extent), 99% were ever smokers at baseline (active and former smokers combined) compared to 42% in the 4th (highest) quartile (>62% methylation extent). For former smokers, the cumulative incidence of myocardial infarction was higher in the lowest methylation extent (1st-50th percentile) compared to the highest methylation extent (51st-100th percentile). Compared to never smokers, the multivariable adjusted subhazard ratio for myocardial infarction was 1.09 (95%CI:0.88-1.35) for former smokers with the highest methylation degree, 1.38 (1.06-1.80) for active smokers with the highest methylation extent, 1.39 (1.08-1.78) for former smokers with the lowest methylation extent, and 1.61 (1.35-1.92) for active smokers with the lowest methylation extent. CONCLUSIONS AHRR cg05575921 hypomethylation as an epigenetic marker of smoking history predicts risk of myocardial infarction, particularly in former smokers. Further, AHRR hypomethylation, regardless of smoking status, was associated with increased risk of myocardial infarction.
Collapse
|
46
|
Kumar P, Yadav M, Verma K, Dixit R, Singh J, Tiwary SK, Narayan G, Dixit VK. Expression analysis of aryl hydrocarbon receptor repressor (AHRR) gene in gallbladder cancer. Saudi J Gastroenterol 2020; 27:54-59. [PMID: 32801256 PMCID: PMC8083244 DOI: 10.4103/sjg.sjg_213_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND The aryl hydrocarbon receptor repressor (AHRR), a member of the growing superfamily, is a basic helix-loop-helix/PerAHR nuclear translocator (ARNT)-Sim (bHLH-PAS) protein. AHRR has been proposed to function as a putative new tumor suppressor gene based on studies in multiple types of human cancers. This current study aims to investigate AHHR expression and its prognostic significance in gallbladder cancer. METHODS The study includes 48 gallbladder cancer and 34 chronic cholecystitis cases as controls. The expression level of AHRR was analyzed by using semi-quantitative PCR and immunohistochemical staining. The results were correlated with different clinical parameters. RESULTS We demonstrate that the expression of AHRR is significantly down-regulated in gallbladder cancer tissue samples as compared to that in chronic cholecystitis tissue samples by reverse transcriptase PCR (RT-PCR) (P = 0.017) and immunohistochemistry analysis (P = 0.002). Interestingly, our RT-PCR data revealed that AHRR mRNA expression is frequently down-regulated (45.8%; 22/48) in cases as compared to 14.7% (5/34) in controls. Similarly, immunohistochemical analysis data show significant down-regulation of AHRR expression in 77.1% (37/48) of gallbladder cancer cases than 44.1% (15/34) in controls (P < 0.017). Reduced mRNA and protein expression is significantly associated with advanced T-stage (P = 0.001), histological differentiation (P = 0.001), and tumors with nodal metastasis (P = 0.001). Decreased expression of AHRR is significantly associated with poor prognosis in gallbladder cancer patients. CONCLUSION In conclusion, the present study suggests that low AHRR expression may be critical in gallbladder cancer development. Our data suggests that AHRR may act as a tumor suppressor gene and its expression profile may be useful as a diagnostic marker in gallbladder cancer.
Collapse
Affiliation(s)
- Puneet Kumar
- Department of Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India,Address for correspondence: Prof. Puneet Kumar, Professor and Head, Department of Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi - 221 005, Uttar Pradesh, India. E-mail:
| | - Manoj Yadav
- Department of Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Khushi Verma
- Department of Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ruhi Dixit
- Department of Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Juhi Singh
- Department of Gastroenterology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Satyendra K Tiwary
- Department of Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Gopeshwar Narayan
- Department of Molecular and Human Genetics, Faculty of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - V K Dixit
- Department of Gastroenterology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
47
|
Tantoh DM, Wu MC, Chuang CC, Chen PH, Tyan YS, Nfor ON, Lu WY, Liaw YP. AHRR cg05575921 methylation in relation to smoking and PM 2.5 exposure among Taiwanese men and women. Clin Epigenetics 2020; 12:117. [PMID: 32736658 PMCID: PMC7394684 DOI: 10.1186/s13148-020-00908-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
Background Polycyclic aromatic hydrocarbon (PAH)-rich substances like cigarette smoke and PM2.5 induce aryl hydrocarbon receptor (AHR)-mediated aryl hydrocarbon receptor repressor (AHRR) methylation. AHRR cg05575921 and coagulation factor II (thrombin) receptor-like 3 (F2RL3) cg03636183 methylation patterns are well-established biomarkers for smoking. Even though AHRR cg05575921 methylation has recently been associated with PM2.5, the interaction between smoking and PM2.5 on AHRR methylation is yet to be fully explored. We evaluated AHRR and F2RL3 CpG sites to identify potential significant markers in relation to PM2.5 and smoking in Taiwanese adults. Methods DNA methylation and smoking data of 948 participants aged 30–70 years were obtained from the Taiwan Biobank Database (2008–2015), while PM2.5 data were obtained from the Air Quality Monitoring Database (2006–2011). Results Smoking and PM2.5 were independently associated with hypomethylation (lower levels) of AHRR cg05575921, AHRR cg23576855, F2RL3 cg03636183, and F2LR3 cg21911711 after multiple-comparison correction (Bonferroni P < 0.00028409). Cg05575921 was the most hypomethylated AHRR CpG site, while cg03636183 was the most hypomethylated F2RL3 CpG site. Overall, cg05575921 was the most hypomethylated CpG site: β = − 0.03909, P < 0.0001; − 0.17536, P < 0.0001 for former and current smoking, respectively (P-trendsmoking < 0.0001) and − 0.00141, P < 0.0001 for PM2.5. After adjusting for F2RL3 cg03636183, smoking and PM2.5 remained significantly associated with cg05575921 hypomethylation: β − 0.02221, P < 0.0001; − 0.11578, P < 0.0001 for former and current smoking, respectively (P-trendsmoking < 0.0001) and − 0.0070, P = 0.0120 for PM2.5. After stratification by sex, smoking and PM2.5 remained associated (P < 0.05) with cg05575921 hypomethylation in both men (β = − 0.04274, − 0.17700, and − 0.00163 for former smoking, current smoking, and PM2.5, respectively) and women (β = − 0.01937, − 0.17255, and − 0.00105 for former smoking, current smoking, and PM2.5, respectively). After stratification by residential area, former and current smoking remained associated (P < 0.05) with cg05575921 hypomethylation: β = − 0.03918 and − 0.17536, respectively (P-trendsmoking < 0.0001). Living in the central and southern areas was also associated (P < 0.05) with cg05575921 hypomethylation: β = − 0.01356 and − 0.01970, respectively (P-trendarea < 0.0001). Conclusion Smoking and PM2.5 were independently associated with hypomethylation of cg05575921, cg23576855, cg03636183, and cg21911711. The most hypomethylated CpG site was cg05575921 and its association with smoking and PM2.5 was dose-dependent.
Collapse
Affiliation(s)
- Disline Manli Tantoh
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, Taiwan.,Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
| | - Ming-Chi Wu
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung City, Taiwan.,School of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung City, Taiwan.,School of Medical Informatics, Chung Shan Medical University, Taichung City, 40201, Taiwan
| | - Chun-Chao Chuang
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, Taiwan.,School of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung City, Taiwan
| | - Pei-Hsin Chen
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
| | - Yeu Sheng Tyan
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung City, Taiwan.,School of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung City, Taiwan.,Medical Imaging and Big Data Center, Chung Shan Medical University Hospital, Taichung City, Taiwan
| | - Oswald Ndi Nfor
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
| | - Wen-Yu Lu
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
| | - Yung-Po Liaw
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, Taiwan. .,Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan. .,Medical Imaging and Big Data Center, Chung Shan Medical University Hospital, Taichung City, Taiwan.
| |
Collapse
|
48
|
Ringh MV, Hagemann-Jensen M, Needhamsen M, Kullberg S, Wahlström J, Grunewald J, Brynedal B, Jagodic M, Ekström TJ, Öckinger J, Kular L. Methylome and transcriptome signature of bronchoalveolar cells from multiple sclerosis patients in relation to smoking. Mult Scler 2020; 27:1014-1026. [PMID: 32729352 PMCID: PMC8145441 DOI: 10.1177/1352458520943768] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Despite compelling evidence that cigarette smoking impacts the risk of developing multiple sclerosis (MS), little is known about smoking-associated changes in the primary exposed lung cells of patients. OBJECTIVES We aimed to examine molecular changes occurring in bronchoalveolar lavage (BAL) cells from MS patients in relation to smoking and in comparison to healthy controls (HCs). METHODS We profiled DNA methylation in BAL cells from female MS (n = 17) and HC (n = 22) individuals, using Illumina Infinium EPIC and performed RNA-sequencing in non-smokers. RESULTS The most prominent changes were found in relation to smoking, with 1376 CpG sites (adjusted P < 0.05) differing between MS smokers and non-smokers. Approximately 30% of the affected genes overlapped with smoking-associated changes in HC, leading to a strong common smoking signature in both MS and HC after gene ontology analysis. Smoking in MS patients resulted in additional discrete changes related to neuronal processes. Methylome and transcriptome analyses in non-smokers suggest that BAL cells from MS patients display very subtle (not reaching adjusted P < 0.05) but concordant changes in genes connected to reduced transcriptional/translational processes and enhanced cellular motility. CONCLUSIONS Our study provides insights into the impact of smoking on lung inflammation and immunopathogenesis of MS.
Collapse
Affiliation(s)
- Mikael V Ringh
- Department of Clinical Neuroscience and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Michael Hagemann-Jensen
- Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maria Needhamsen
- Department of Clinical Neuroscience and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Susanna Kullberg
- Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden/Respiratory Medicine Division, Department of Medicine and Theme Inflammation and Infection, Karolinska University Hospital, Stockholm, Sweden
| | - Jan Wahlström
- Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johan Grunewald
- Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Boel Brynedal
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tomas J Ekström
- Department of Clinical Neuroscience and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johan Öckinger
- Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lara Kular
- Department of Clinical Neuroscience and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
49
|
Current status of development of methylation biomarkers for in vitro diagnostic IVD applications. Clin Epigenetics 2020; 12:100. [PMID: 32631437 PMCID: PMC7336678 DOI: 10.1186/s13148-020-00886-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
A significant volume of research clearly shows that disease-related methylation changes can be used as biomarkers at all stages of clinical disease management, including risk assessment and predisposition screening through early diagnostics to personalization of patient care and monitoring of the relapse and chronic disease. Thus disease-related methylation changes are an attractive source of the biomarkers that can have significant impact on precision medicine. However, the translation of the research findings in methylation biomarkers field to clinical practice is at the very least not satisfactory. That is mainly because the evidence generated in research studies indicating the utility of the disease-related methylation change to predict clinical outcome is in majority of the cases not sufficient to postulate the diagnostic use of the biomarker. The research studies need to be followed by well-designed and systematic investigations of clinical utility of the biomarker that produce data of sufficient quality to meet regulatory approval for the test to be used to make clinically valid decision. In this review, we describe methylation-based IVD tests currently approved for IVD use or at the advanced stages of the development for the diagnostic use. For each of those tests, we analyze the technologies that the test utilizes for methylation detection as well as describe the types of the clinical studies that were performed to show clinical validity of the test and warrant regulatory approval. The examples reviewed here should help with planning of clinical investigations and delivery of the clinical evidence required for the regulatory approval of potential methylation biomarker based IVD tests.
Collapse
|
50
|
Philibert R, Beach SR, Lei MK, Gibbons FX, Gerrard M, Simons RL, Dogan MV. Array-Based Epigenetic Aging Indices May Be Racially Biased. Genes (Basel) 2020; 11:genes11060685. [PMID: 32580526 PMCID: PMC7349894 DOI: 10.3390/genes11060685] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/01/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022] Open
Abstract
Epigenetic aging (EA) indices are frequently used as predictors of mortality and other important health outcomes. However, each of the commonly used array-based indices has significant heritable components which could tag ethnicity and potentially confound comparisons across racial and ethnic groups. To determine if this was possible, we examined the relationship of DNA methylation in cord blood from 203 newborns (112 African American (AA) and 91 White) at the 513 probes from the Levine PhenoAge Epigenetic Aging index to ethnicity. Then, we examined all sites significantly associated with race in the newborn sample to determine if they were also associated with an index of ethnic genetic heritage in a cohort of 505 AA adults. After Bonferroni correction, methylation at 50 CpG sites was significantly associated with ethnicity in the newborn cohort. The five most significant sites predicted ancestry with a receiver operator characteristic area under the curve of 0.97. Examination of the top 50 sites in the AA adult cohort showed that methylation status at 11 of those sites was also associated with percentage European ancestry. We conclude that the Levine PhenoAge Index is influenced by cryptic ethnic-specific genetic influences. This influence may extend to similarly constructed EA indices and bias cross-race comparisons.
Collapse
Affiliation(s)
- Robert Philibert
- Department of Psychiatry, University of Iowa, Iowa City, IA 52242, USA;
- Behavioral Diagnostics LLC, Coralville, IA 52241, USA
- Correspondence: ; Tel.: +1-319-353-4986
| | - Steven R.H. Beach
- Center for Family Research, University of Georgia, Athens, GA 30602, USA; (S.R.H.B.); (M.-K.L.); (R.L.S.)
| | - Man-Kit Lei
- Center for Family Research, University of Georgia, Athens, GA 30602, USA; (S.R.H.B.); (M.-K.L.); (R.L.S.)
| | - Frederick X. Gibbons
- Department of Psychological Sciences, University of Connecticut, Storrs, CT 06268, USA; (F.X.G.); (M.G.)
| | - Meg Gerrard
- Department of Psychological Sciences, University of Connecticut, Storrs, CT 06268, USA; (F.X.G.); (M.G.)
| | - Ronald L. Simons
- Center for Family Research, University of Georgia, Athens, GA 30602, USA; (S.R.H.B.); (M.-K.L.); (R.L.S.)
| | | |
Collapse
|