1
|
Pośpiech E, Rudnicka J, Noroozi R, Pisarek-Pacek A, Wysocka B, Masny A, Boroń M, Migacz-Gruszka K, Pruszkowska-Przybylska P, Kobus M, Lisman D, Zielińska G, Cytacka S, Iljin A, Wiktorska JA, Michalczyk M, Kaczka P, Krzysztofik M, Sitek A, Spólnicka M, Ossowski A, Branicki W. DNA methylation at AHRR as a master predictor of smoke exposure and a biomarker for sleep and exercise. Clin Epigenetics 2024; 16:147. [PMID: 39425209 PMCID: PMC11490037 DOI: 10.1186/s13148-024-01757-0] [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: 03/22/2024] [Accepted: 10/01/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND DNA methylation profiling may provide a more accurate measure of the smoking status than self-report and may be useful in guiding clinical interventions and forensic investigations. In the current study, blood DNA methylation profiles of nearly 800 Polish individuals were assayed using Illuminia EPIC and the inference of smoking from epigenetic data was explored. In addition, we focused on the role of the AHRR gene as a top marker for smoking and investigated its responsiveness to other lifestyle behaviors. RESULTS We found > 450 significant CpGs associated with cigarette consumption, and overrepresented in various biological functions including cell communication, response to stress, blood vessel development, cell death, and atherosclerosis. The model consisting of cg05575921 in AHRR (p = 4.5 × 10-32) and three additional CpGs (cg09594361, cg21322436 in CNTNAP2 and cg09842685) was able to predict smoking status with a high accuracy of AUC = 0.8 in the test set. Importantly, a gradual increase in the probability of smoking was observed, starting from occasional smokers to regular heavy smokers. Furthermore, former smokers displayed the intermediate DNA methylation profiles compared to current and never smokers, and thus our results indicate the potential reversibility of DNA methylation after smoking cessation. The AHRR played a key role in a predictive analysis, explaining 21.5% of the variation in smoking. In addition, the AHRR methylation was analyzed for association with other modifiable lifestyle factors, and showed significance for sleep and physical activity. We also showed that the epigenetic score for smoking was significantly correlated with most of the epigenetic clocks tested, except for two first-generation clocks. CONCLUSIONS Our study suggests that a more rapid return to never-smoker methylation levels after smoking cessation may be achievable in people who change their lifestyle in terms of physical activity and sleep duration. As cigarette smoking has been implicated in the literature as a leading cause of epigenetic aging and AHRR appears to be modifiable by multiple exogenous factors, it emerges as a promising target for intervention and investment.
Collapse
Affiliation(s)
- Ewelina Pośpiech
- Department of Forensic Genetics, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland.
| | - Joanna Rudnicka
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Rezvan Noroozi
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
- Johns Hopkins University School of Medicine, Baltimore, USA
| | - Aleksandra Pisarek-Pacek
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
- Institute of Zoology and Biomedical Research of the Jagiellonian University, Krakow, Poland
| | - Bożena Wysocka
- Central Forensic Laboratory of the Police, Warsaw, Poland
| | | | - Michał Boroń
- Central Forensic Laboratory of the Police, Warsaw, Poland
| | | | | | - Magdalena Kobus
- Institute of Biological Sciences, Faculty of Biology and Environmental Sciences, Cardinal Stefan Wyszynski University in Warsaw, Warsaw, Poland
| | - Dagmara Lisman
- Department of Forensic Genetics, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Grażyna Zielińska
- Department of Forensic Genetics, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Sandra Cytacka
- Department of Forensic Genetics, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Aleksandra Iljin
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Lodz, Lodz, Poland
| | | | - Małgorzata Michalczyk
- Department of Sport Nutrition, The Jerzy Kukuczka Academy of Physical Education in Katowice, Katowice, Poland
| | - Piotr Kaczka
- Department of Sport Nutrition, The Jerzy Kukuczka Academy of Physical Education in Katowice, Katowice, Poland
| | - Michał Krzysztofik
- Institute of Sports Sciences, The Jerzy Kukuczka Academy of Physical Education in Katowice, Katowice, Poland
| | - Aneta Sitek
- Department of Anthropology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | | | - Andrzej Ossowski
- Department of Forensic Genetics, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Wojciech Branicki
- Institute of Zoology and Biomedical Research of the Jagiellonian University, Krakow, Poland
- Institute of Forensic Research, Krakow, Poland
| |
Collapse
|
2
|
Meeks GL, Scelza B, Asnake HM, Prall S, Patin E, Froment A, Fagny M, Quintana-Murci L, Henn BM, Gopalan S. Common DNA sequence variation influences epigenetic aging in African populations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.608843. [PMID: 39253488 PMCID: PMC11383046 DOI: 10.1101/2024.08.26.608843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Aging is associated with genome-wide changes in DNA methylation in humans, facilitating the development of epigenetic age prediction models. However, most of these models have been trained primarily on European-ancestry individuals, and none account for the impact of methylation quantitative trait loci (meQTL). To address these gaps, we analyzed the relationships between age, genotype, and CpG methylation in 3 understudied populations: central African Baka (n = 35), southern African ‡Khomani San (n = 52), and southern African Himba (n = 51). We find that published prediction methods yield higher mean errors in these cohorts compared to European-ancestry individuals, and find that unaccounted-for DNA sequence variation may be a significant factor underlying this loss of accuracy. We leverage information about the associations between DNA genotype and CpG methylation to develop an age predictor that is minimally influenced by meQTL, and show that this model remains accurate across a broad range of genetic backgrounds. Intriguingly, we also find that the older individuals and those exhibiting relatively lower epigenetic age acceleration in our cohorts tend to carry more epigenetic age-reducing genetic variants, suggesting a novel mechanism by which heritable factors can influence longevity.
Collapse
Affiliation(s)
- Gillian L. Meeks
- Integrative Genetics and Genomics Graduate Program, University of California, Davis, CA 95694, USA
| | - Brooke Scelza
- Department of Anthropology, University of California, Los Angeles, CA, 90095, USA
| | - Hana M. Asnake
- Forensic Science Graduate Program, University of California, Davis, CA, 95694, USA
| | - Sean Prall
- Department of Anthropology, University of California, Los Angeles, CA, 90095, USA
| | - Etienne Patin
- Human Evolutionary Genetics Unit, CNRS UMR2000, Paris, 75015, France
| | - Alain Froment
- Institut de Recherche pour le Développement, UMR 208, Muséum National d’Histoire Naturelle, Paris, 75005, France
| | - Maud Fagny
- Human Evolutionary Genetics Unit, CNRS UMR2000, Paris, 75015, France
- Université Paris-Saclay, INRAE, CNRS, AgroParisTech, Genetique Quantitative et Evolution - Le Moulon, Gif-sur-Yvette, 91190, France
| | | | - Brenna M. Henn
- Department of Anthropology, University of California Davis, Davis, CA, 95616, USA
- UC Davis Genome Center and Center for Population Biology, University of California, Davis, CA 95694, USA
| | - Shyamalika Gopalan
- Department of Ecology and Evolution, Stony Brook University, Stony Brook, NY, 11790, USA
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA
- Center for Human Genetics, Clemson University, Greenwood, SC 29646, USA
| |
Collapse
|
3
|
Clark SL, McGinnis EW, Zhao M, Xie L, Marks GT, Aberg KA, van den Oord EJCG, Copeland WE. The Impact of Childhood Mental Health and Substance Use on Methylation Aging Into Adulthood. J Am Acad Child Adolesc Psychiatry 2024; 63:825-834. [PMID: 38157979 DOI: 10.1016/j.jaac.2023.10.014] [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: 09/12/2022] [Revised: 10/11/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVE To test whether childhood mental health symptoms, substance use, and early adversity accelerate the rate of DNA methylation (DNAm) aging from adolescence to adulthood. METHOD DNAm was assayed from blood samples in 381 participants in both adolescence (mean [SD] age = 13.9 [1.6] years) and adulthood (mean [SD] age = 25.9 [2.7] years). Structured diagnostic interviews were completed with participants and their parents at multiple childhood observations (1,950 total) to assess symptoms of common mental health disorders (attention-deficit/hyperactivity disorder, oppositional defiant disorder, conduct disorder, anxiety, and depression) and common types of substance use (alcohol, cannabis, nicotine) and early adversities. RESULTS Neither childhood mental health symptoms nor substance use variables were associated with DNAm aging cross-sectionally. In contrast, the following mental health symptoms and substance variables were associated with accelerated DNAm aging from adolescence to adulthood: depressive symptoms (b = 0.314, SE = 0.127, p = .014), internalizing symptoms (b = 0.108, SE = 0.049, p = .029), weekly cannabis use (b =1.665, SE = 0.591, p = .005), and years of weekly cannabis use (b = 0.718, SE = 0.283, p = .012). In models testing all individual variables simultaneously, the combined effect of the variables was equivalent to a potential difference of 3.17 to 3.76 years in DNAm aging. A final model tested a variable assessing cumulative exposure to mental health symptoms, substance use, and early adversities. This cumulative variable was strongly associated with accelerated aging (b = 0.126, SE = 0.044, p = .005). CONCLUSION Mental health symptoms and substance use accelerated DNAm aging into adulthood in a manner consistent with a shared risk mechanism. PLAIN LANGUAGE SUMMARY Using data from 381 participants in the Great Smoky Mountains Study, the authors examined whether childhood mental health symptoms, substance use, and early adversity accelerate biological aging, as measured by DNA methylation age, from adolescence to adulthood. Depressive symptoms and cannabis use were found to significantly accelerate biological aging. Models that tested the combined effect of mental health symptoms, substance use, and early adversity demonstrated that there was a shared effect across these types of childhood problems on accelerated aging.
Collapse
Affiliation(s)
| | | | - Min Zhao
- Virginia Commonwealth University, Richmond, Virginia
| | - Linying Xie
- Virginia Commonwealth University, Richmond, Virginia
| | | | | | | | | |
Collapse
|
4
|
Klopack ET, Crimmins EM. Epigenetic Aging Helps Explain Differential Resilience in Older Adults. Demography 2024; 61:1023-1041. [PMID: 39012228 PMCID: PMC11485224 DOI: 10.1215/00703370-11466635] [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] [Indexed: 07/17/2024]
Abstract
Past research suggests that resilience to health hazards increases with age, potentially because less resilient individuals die at earlier ages, leaving behind their more resilient peers. Using lifetime cigarette smoking as a model health hazard, we examined whether accelerated epigenetic aging (indicating differences in the speed of individuals' underlying aging process) helps explain age-related resilience in a nationally representative sample of 3,783 older U.S. adults from the Health and Retirement Study. Results of mediation moderation analyses indicated that participants aged 86 or older showed a weaker association between lifetime cigarette smoking and mortality relative to participants aged 76-85 and a weaker association between smoking and multimorbidity relative to all younger cohorts. This moderation effect was mediated by a reduced association between smoking pack-years and epigenetic aging. This research helps identify subpopulations of particularly resilient individuals and identifies epigenetic aging as a potential mechanism explaining this process. Interventions in younger adults could utilize epigenetic aging estimates to identify the most vulnerable individuals and intervene before adverse health outcomes, such as chronic disease morbidity or mortality, manifest.
Collapse
Affiliation(s)
- Eric T Klopack
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Eileen M Crimmins
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
5
|
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
|
6
|
Sehgal R, Markov Y, Qin C, Meer M, Hadley C, Shadyab AH, Casanova R, Manson JE, Bhatti P, Crimmins EM, Hägg S, Assimes TL, Whitsel EA, Higgins-Chen AT, Levine M. Systems Age: A single blood methylation test to quantify aging heterogeneity across 11 physiological systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.13.548904. [PMID: 37503069 PMCID: PMC10370047 DOI: 10.1101/2023.07.13.548904] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Individuals, organs, tissues, and cells age in diverse ways throughout the lifespan. Epigenetic clocks attempt to quantify differential aging between individuals, but they typically summarize aging as a single measure, ignoring within-person heterogeneity. Our aim was to develop novel systems-based methylation clocks that, when assessed in blood, capture aging in distinct physiological systems. We combined supervised and unsupervised machine learning methods to link DNA methylation, system-specific clinical chemistry and functional measures, and mortality risk. This yielded a panel of 11 system-specific scores- Heart, Lung, Kidney, Liver, Brain, Immune, Inflammatory, Blood, Musculoskeletal, Hormone, and Metabolic. Each system score predicted a wide variety of outcomes, aging phenotypes, and conditions specific to the respective system. We also combined the system scores into a composite Systems Age clock that is predictive of aging across physiological systems in an unbiased manner. Finally, we showed that the system scores clustered individuals into unique aging subtypes that had different patterns of age-related disease and decline. Overall, our biological systems based epigenetic framework captures aging in multiple physiological systems using a single blood draw and assay and may inform the development of more personalized clinical approaches for improving age-related quality of life.
Collapse
|
7
|
Lin F, Chen X, Shi Y, Yang K, Hu G, Zhuang W, Lin Y, Huang T, Ye Q, Cai G, Wu X. Early-life tobacco smoke exposure and stroke risk: a prospective study of 341,783 and 352,737 UK Biobank participants. BMC Public Health 2024; 24:1339. [PMID: 38760724 PMCID: PMC11102258 DOI: 10.1186/s12889-024-18588-6] [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: 01/06/2024] [Accepted: 04/14/2024] [Indexed: 05/19/2024] Open
Abstract
INTRODUCTION Stroke is a life-threatening condition that causes a major medical burden globally. The currently used methods for the prevention or prediction of stroke have certain limitations. Exposure to tobacco in early life, including smoking during adolescence and maternal smoking during pregnancy, can affect adolescent development and lead to several negative outcomes. However, the association between early-life tobacco exposure and stroke is not known. METHODS In this prospective cohort study, for the analyses involving exposure to maternal smoking during pregnancy and age of smoking initiation, we included 304,984 and 342,893 participants, respectively., respectively from the UK Biobank. Cox proportional hazard regression model and subgroup analyses were performed to investigate the association between early-life tobacco exposure and stroke. Mediation analyses were performed to identify the mediating role of biological aging in the association between early tobacco exposure and stroke. RESULTS Compared with participants whose mothers did not smoke during pregnancy, participants whose mothers smoked during pregnancy showed an 11% increased risk of stroke (HR: 1.11, 95% CI: 1.05-1.18, P < 0.001). Compared with participants who never smoked, participants who smoked during adulthood, adolescence and childhood showed a 22%, 24%, and 38% increased risk of stroke during their adulthood, respectively. Mediation analysis indicated that early-life tobacco exposure can cause stroke by increasing biological aging. CONCLUSION This study reveals that exposure to tobacco during early life is associated with an increased risk of experiencing a stroke, and increased biological aging can be the underlying mechanism.
Collapse
Affiliation(s)
- Fabin Lin
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, 350001, Fuzhou, China
- Fujian Medical University, Fuzhou, China
- Department of Neurosurgery, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
| | - Xuanjie Chen
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, 350001, Fuzhou, China
- Fujian Medical University, Fuzhou, China
| | - Yisen Shi
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, 350001, Fuzhou, China
- Fujian Medical University, Fuzhou, China
| | - Kaitai Yang
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, 350001, Fuzhou, China
- Fujian Medical University, Fuzhou, China
| | - Guoping Hu
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, 350001, Fuzhou, China
- Fujian Medical University, Fuzhou, China
| | - Weijiang Zhuang
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, 350001, Fuzhou, China
- Fujian Medical University, Fuzhou, China
| | - Yifei Lin
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, 350001, Fuzhou, China
- Fujian Medical University, Fuzhou, China
| | - Tingting Huang
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, 350001, Fuzhou, China
- Fujian Medical University, Fuzhou, China
| | - Qinyong Ye
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China.
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, 350001, Fuzhou, China.
- Fujian Medical University, Fuzhou, China.
| | - Guoen Cai
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China.
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, 350001, Fuzhou, China.
- Fujian Medical University, Fuzhou, China.
| | - Xilin Wu
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China.
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, 350001, Fuzhou, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, 350001, Fuzhou, China.
- Fujian Medical University, Fuzhou, China.
| |
Collapse
|
8
|
Ambroa-Conde A, Casares de Cal MA, Gómez-Tato A, Robinson O, Mosquera-Miguel A, de la Puente M, Ruiz-Ramírez J, Phillips C, Lareu MV, Freire-Aradas A. Inference of tobacco and alcohol consumption habits from DNA methylation analysis of blood. Forensic Sci Int Genet 2024; 70:103022. [PMID: 38309257 DOI: 10.1016/j.fsigen.2024.103022] [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: 08/22/2023] [Revised: 12/22/2023] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
DNA methylation has become a biomarker of great interest in the forensic and clinical fields. In criminal investigations, the study of this epigenetic marker has allowed the development of DNA intelligence tools providing information that can be useful for investigators, such as age prediction. Following a similar trend, when the origin of a sample in a criminal scenario is unknown, the inference of an individual's lifestyle such as tobacco use and alcohol consumption could provide relevant information to help in the identification of DNA donors at the crime scene. At the same time, in the clinical domain, prediction of these trends of consumption could allow the identification of people at risk or better identification of the causes of different pathologies. In the present study, DNA methylation data from the UK AIRWAVE study was used to build two binomial logistic models for the inference of smoking and drinking status. A total of 348 individuals (116 non-smokers, 116 former smokers and 116 smokers) plus a total of 237 individuals (79 non-drinkers, 79 moderate drinkers and 79 drinkers) were used for development of tobacco and alcohol consumption prediction models, respectively. The tobacco prediction model was composed of two CpGs (cg05575921 in AHRR and cg01940273) and the alcohol prediction model three CpGs (cg06690548 in SLC7A11, cg0886875 and cg21294714 in MIR4435-2HG), providing correct classifications of 86.49% and 74.26%, respectively. Validation of the models was performed using leave-one-out cross-validation. Additionally, two independent testing sets were also assessed for tobacco and alcohol consumption. Considering that the consumption of these substances could underlie accelerated epigenetic ageing patterns, the effect of these lifestyles on the prediction of age was evaluated. To do that, a quantile regression model based on previous studies was generated, and the potential effect of tobacco and alcohol consumption with the epigenetic age was assessed. The Wilcoxon test was used to evaluate the residuals generated by the model and no significant differences were observed between the categories analyzed.
Collapse
Affiliation(s)
- A Ambroa-Conde
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - M A Casares de Cal
- CITMAga (Center for Mathematical Research and Technology of Galicia), University of Santiago de Compostela, Spain
| | - A Gómez-Tato
- CITMAga (Center for Mathematical Research and Technology of Galicia), University of Santiago de Compostela, Spain
| | - O Robinson
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - A Mosquera-Miguel
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - M de la Puente
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - J Ruiz-Ramírez
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - C Phillips
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - M V Lareu
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - A Freire-Aradas
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain.
| |
Collapse
|
9
|
Chen H, Tang H, Zhang X, Huang J, Luo N, Guo Q, Wang X. Adherence to Life's Essential 8 is associated with delayed biological aging: a population-based cross-sectional study. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2024:S1885-5857(24)00142-7. [PMID: 38663840 DOI: 10.1016/j.rec.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/10/2024] [Indexed: 06/06/2024]
Abstract
INTRODUCTION AND OBJECTIVES The aim of this study was to explore the potential of adhering to the American Heart Association's updated Life's Essential 8 (LE8) scores in delaying biological aging amid growing concerns about aging populations and related diseases. METHODS A total of 18 261 adults (≥ 20 years old) were examined using National Health and Nutrition Examination Survey data from 2005-2010 and 2015-2018. The LE8 includes 8 components, covering health behaviors and factors. Acceleration of biological aging was defined as an excess of biological/phenotypic age over chronological age, assessed by using clinical biomarkers. The association between LE8 score and biological aging was explored through regression analyses. RESULTS Each 10-point increase in LE8 scores was associated with a 1.19-year decrease in biological age and a 1.63-year decrease in phenotypic age. Individuals with high cardiovascular health (CVH) had a 90% reduction in their risk of accelerated aging based on biological age and an 81% reduction based on phenotypic age compared with individuals with low CVH. Bootstrap-based model estimates and weighted quantile sum regression suggested that health factors, particularly blood glucose, had strong impact on delaying aging. The association between smoking and biological aging seemed to differ depending on the definition of aging used. Among all subgroups, LE8 consistently correlated negatively with biological aging, despite observed interactions. Three sensitivity analyses confirmed the robustness of our conclusions. CONCLUSIONS A higher CVH is associated with a lower risk of biological aging. Maintaining elevated LE8 levels across demographics, regardless of cardiovascular history, is recommended to delay aging and promote healthy aging, with significant implications for primary health care.
Collapse
Affiliation(s)
- Hongyu Chen
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Haoxian Tang
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China; Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Xuan Zhang
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jingtao Huang
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China; Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Nan Luo
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China; Department of Psychiatry, Shantou University Mental Health Center, Shantou, China
| | - Qingqian Guo
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Xin Wang
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China.
| |
Collapse
|
10
|
Zheng Y, Lunetta KL, Liu C, Smith AK, Sherva R, Miller MW, Logue MW. A novel principal component based method for identifying differentially methylated regions in Illumina Infinium MethylationEPIC BeadChip data. Epigenetics 2023; 18:2207959. [PMID: 37196182 PMCID: PMC10193914 DOI: 10.1080/15592294.2023.2207959] [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: 09/18/2022] [Revised: 03/22/2023] [Accepted: 04/19/2023] [Indexed: 05/19/2023] Open
Abstract
Differentially methylated regions (DMRs) are genomic regions with methylation patterns across multiple CpG sites that are associated with a phenotype. In this study, we proposed a Principal Component (PC) based DMR analysis method for use with data generated using the Illumina Infinium MethylationEPIC BeadChip (EPIC) array. We obtained methylation residuals by regressing the M-values of CpGs within a region on covariates, extracted PCs of the residuals, and then combined association information across PCs to obtain regional significance. Simulation-based genome-wide false positive (GFP) rates and true positive rates were estimated under a variety of conditions before determining the final version of our method, which we have named DMRPC. Then, DMRPC and another DMR method, coMethDMR, were used to perform epigenome-wide analyses of several phenotypes known to have multiple associated methylation loci (age, sex, and smoking) in a discovery and a replication cohort. Among regions that were analysed by both methods, DMRPC identified 50% more genome-wide significant age-associated DMRs than coMethDMR. The replication rate for the loci that were identified by only DMRPC was higher than the rate for those that were identified by only coMethDMR (90% for DMRPC vs. 76% for coMethDMR). Furthermore, DMRPC identified replicable associations in regions of moderate between-CpG correlation which are typically not analysed by coMethDMR. For the analyses of sex and smoking, the advantage of DMRPC was less clear. In conclusion, DMRPC is a new powerful DMR discovery tool that retains power in genomic regions with moderate correlation across CpGs.
Collapse
Affiliation(s)
- Yuanchao Zheng
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Chunyu Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Alicia K. Smith
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Richard Sherva
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Mark W. Miller
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, USA
- Biomedical Genetics, Boston University School of Medicine, Boston, MA, USA
| | - Mark W. Logue
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
- Biomedical Genetics, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
11
|
Gao S, Deng H, Wen S, Wang Y. Effects of accelerated biological age on depressive symptoms in a causal reasoning framework. J Affect Disord 2023; 339:732-741. [PMID: 37442448 DOI: 10.1016/j.jad.2023.07.019] [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] [Received: 04/09/2023] [Revised: 06/07/2023] [Accepted: 07/08/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND Depression in middle-aged and elderly individuals is multifaceted and heterogeneous, linked to biological age (BA) based on aging-related biomarkers. However, due to confounding with chronological age and the absence of subgroup analysis and causal reasoning, the association between BA and depressive symptoms (DS) might be unstable and requires further investigation. METHODS We utilized data from the China Health and Retirement Longitudinal Study (N = 9478) to perform association analysis, causal inference, and subgroup analysis. BA acceleration (BAA) was derived using machine learning and adjusted for chronological age. A generalized linear mixed-effects model (GLMM) tree algorithm was employed to identify subgroups. The causal reasoning frame included propensity score matching and fast large-scale almost matching exactly. RESULTS In the longitudinal analysis, BAA exhibited a consistent and significant positive association with DS, even after controlling for demographic characteristics, lifestyle factors, health status, and physical functions. This association remained unchanged within the causal framework. GLMM tree analysis identified three partitioning variables (sex, satisfaction, and BMI) and five subgroups. Further subgroup analysis revealed that BAA exerted the strongest effect on DS among women with less satisfying lives. LIMITATIONS Depressive symptoms were evaluated through scale measurements rather than clinical diagnosis. The sample was derived from the general population, not the clinically depressed population. CONCLUSIONS This study provided the first longitudinal evidence that biological age acceleration increases depressive symptoms under causal reasoning and subgroup analysis, particularly among less satisfied women. And the association between BAA and DS was independent of known risk factors.
Collapse
Affiliation(s)
- Sunan Gao
- School of Statistics, Renmin University of China, Beijing, China
| | - Heming Deng
- School of Statistics, Renmin University of China, Beijing, China
| | - Shaobo Wen
- School of Statistics, Renmin University of China, Beijing, China
| | - Yu Wang
- Center for Applied Statistics, Renmin University of China, Beijing, China; School of Statistics, Renmin University of China, Beijing, China.
| |
Collapse
|
12
|
Song MA, Mori KM, McElroy JP, Freudenheim JL, Weng DY, Reisinger SA, Brasky TM, Wewers MD, Shields PG. Accelerated epigenetic age, inflammation, and gene expression in lung: comparisons of smokers and vapers with non-smokers. Clin Epigenetics 2023; 15:160. [PMID: 37821974 PMCID: PMC10568901 DOI: 10.1186/s13148-023-01577-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 10/01/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Cigarette smoking and aging are the main risk factors for pulmonary diseases, including cancer. Epigenetic aging may explain the relationship between smoking, electronic cigarette vaping, and pulmonary health. No study has examined smoking and vaping-related epigenetic aging in relation to lung biomarkers. METHODS Lung epigenetic aging measured by DNA methylation (mAge) and its acceleration (mAA) was assessed in young (age 21-30) electronic cigarette vapers (EC, n = 14, including 3 never-smoking EC), smokers (SM, n = 16), and non-EC/non-SM (NS, n = 39). We investigated relationships of mAge estimates with chronological age (Horvath-mAge), lifespan/mortality (Grim-mAge), telomere length (TL-mAge), smoking/EC history, urinary biomarkers, lung cytokines, and transcriptome. RESULTS Compared to NS, EC and SM had significantly older Grim-mAge, shorter TL-mAge, significantly accelerated Grim-mAge and decelerated TL-mAge. Among SM, Grim-mAA was associated with nicotine intake and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL). For EC, Horvath-mAA was significantly correlated with puffs per day. Overall, cytokines (IL-1β, IL-6, and IL-8) and 759 transcripts (651 unique genes) were significantly associated with Grim-mAA. Grim-mAA-associated genes were highly enriched in immune-related pathways and genes that play a role in the morphology and structures of cells/tissues. CONCLUSIONS Faster lung mAge for SM is consistent with prior studies of blood. Faster lung mAge for EC compared to NS indicates possible adverse pulmonary effects of EC on biological aging. Our findings support further research, particularly on epigenetic markers, on effects of smoking and vaping on pulmonary health. Given that most EC are former smokers, further study is needed to understand unique effects of electronic cigarettes on biological aging.
Collapse
Affiliation(s)
- Min-Ae Song
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, 404 Cunz Hall, 1841 Neil Ave., Columbus, OH, 43210, USA.
| | - Kellie M Mori
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, 404 Cunz Hall, 1841 Neil Ave., Columbus, OH, 43210, USA
| | - Joseph P McElroy
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Jo L Freudenheim
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY, USA
| | - Daniel Y Weng
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH, USA
| | - Sarah A Reisinger
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH, USA
| | - Theodore M Brasky
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH, USA
| | - Mark D Wewers
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH, USA
| | - Peter G Shields
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH, USA
| |
Collapse
|
13
|
Song Y, Liu YS, Talarico F, Zhang Y, Hayward J, Wang M, Stroulia E, Dixon RA, Greiner R, Li X, Greenshaw A, Jie S, Cao B. Associations between Differential Aging and Lifestyle, Environment, Current, and Future Health Conditions: Findings from Canadian Longitudinal Study on Aging. Gerontology 2023; 69:1394-1403. [PMID: 37725932 DOI: 10.1159/000534015] [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: 11/29/2022] [Accepted: 09/05/2023] [Indexed: 09/21/2023] Open
Abstract
INTRODUCTION An aging population will bring a pressing challenge for the healthcare system. Insights into promoting healthy longevity can be gained by quantifying the biological aging process and understanding the roles of modifiable lifestyle and environmental factors, and chronic disease conditions. METHODS We developed a biological age (BioAge) index by applying multiple state-of-art machine learning models based on easily accessible blood test data from the Canadian Longitudinal Study of Aging (CLSA). The BioAge gap, which is the difference between BioAge index and chronological age, was used to quantify the differential aging, i.e., the difference between biological and chronological age, of the CLSA participants. We further investigated the associations between the BioAge gap and lifestyle, environmental factors, and current and future health conditions. RESULTS BioAge gap had strong associations with existing adverse health conditions (e.g., cancers, cardiovascular diseases, diabetes, and kidney diseases) and future disease onset (e.g., Parkinson's disease, diabetes, and kidney diseases). We identified that frequent consumption of processed meat, pork, beef, and chicken, poor outcomes in nutritional risk screening, cigarette smoking, exposure to passive smoking are associated with positive BioAge gap ("older" BioAge than expected). We also identified several modifiable factors, including eating fruits, legumes, vegetables, related to negative BioAge gap ("younger" BioAge than expected). CONCLUSIONS Our study shows that a BioAge index based on easily accessible blood tests has the potential to quantify the differential biological aging process that can be associated with current and future adverse health events. The identified risk and protective factors for differential aging indicated by BioAge gap are informative for future research and guidelines to promote healthy longevity.
Collapse
Affiliation(s)
- Yipeng Song
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada,
| | - Yang S Liu
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
| | - Fernanda Talarico
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
| | - Yanbo Zhang
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
| | - Jake Hayward
- University of Alberta, Department of Emergency Medicine, Edmonton, Alberta, Canada
| | - Mengzhe Wang
- Ministry of Health (Alberta), Edmonton, Alberta, Canada
| | - Eleni Stroulia
- University of Alberta, Department of Computing Science, Edmonton, Alberta, Canada
| | - Roger A Dixon
- University of Alberta, Department of Psychology, Edmonton, Alberta, Canada
| | - Russell Greiner
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
- University of Alberta, Department of Computing Science, Edmonton, Alberta, Canada
| | - Xinmin Li
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
| | - Andrew Greenshaw
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
| | - Sui Jie
- University of Aberdeen, The School of Psychology, Aberdeen, UK
| | - Bo Cao
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
- University of Alberta, Department of Computing Science, Edmonton, Alberta, Canada
| |
Collapse
|
14
|
Vidaki A, Planterose Jiménez B, Poggiali B, Kalamara V, van der Gaag KJ, Maas SCE, Ghanbari M, Sijen T, Kayser M. Targeted DNA methylation analysis and prediction of smoking habits in blood based on massively parallel sequencing. Forensic Sci Int Genet 2023; 65:102878. [PMID: 37116245 DOI: 10.1016/j.fsigen.2023.102878] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/28/2023] [Accepted: 04/18/2023] [Indexed: 04/30/2023]
Abstract
Tobacco smoking is a frequent habit sustained by > 1.3 billion people in 2020 and the leading preventable factor for health risk and premature mortality worldwide. In the forensic context, predicting smoking habits from biological samples may allow broadening DNA phenotyping. In this study, we aimed to implement previously published smoking habit classification models based on blood DNA methylation at 13 CpGs. First, we developed a matching lab tool based on bisulfite conversion and multiplex PCR followed by amplification-free library preparation and targeted paired-end massively parallel sequencing (MPS). Analysis of six technical duplicates revealed high reproducibility of methylation measurements (Pearson correlation of 0.983). Artificially methylated standards uncovered marker-specific amplification bias, which we corrected via bi-exponential models. We then applied our MPS tool to 232 blood samples from Europeans of a wide age range, of which 90 were current, 71 former and 71 never smokers. On average, we obtained 189,000 reads/sample and 15,000 reads/CpG, without marker drop-out. Methylation distributions per smoking category roughly corresponded to previous microarray analysis, showcasing large inter-individual variation but with technology-driven bias. Methylation at 11 out of 13 smoking-CpGs correlated with daily cigarettes in current smokers, while solely one was weakly correlated with time since cessation in former smokers. Interestingly, eight smoking-CpGs correlated with age, and one displayed weak but significant sex-associated methylation differences. Using bias-uncorrected MPS data, smoking habits were relatively accurately predicted using both two- (current/non-current) and three- (never/former/current) category model, but bias correction resulted in worse prediction performance for both models. Finally, to account for technology-driven variation, we built new, joint models with inter-technology corrections, which resulted in improved prediction results for both models, with or without PCR bias correction (e.g. MPS cross-validation F1-score > 0.8; 2-categories). Overall, our novel assay takes us one step closer towards the forensic application of viable smoking habit prediction from blood traces. However, future research is needed towards forensically validating the assay, especially in terms of sensitivity. We also need to further shed light on the employed biomarkers, particularly on the mechanistics, tissue specificity and putative confounders of smoking epigenetic signatures.
Collapse
Affiliation(s)
- Athina Vidaki
- Department of Genetic Identification, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Benjamin Planterose Jiménez
- Department of Genetic Identification, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Brando Poggiali
- Department of Genetic Identification, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Vivian Kalamara
- Department of Genetic Identification, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | | | - Silvana C E Maas
- Department of Genetic Identification, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Titia Sijen
- Division of Biological Traces, Netherlands Forensic Institute, The Hague, the Netherlands; Swammerdam Institute of Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Manfred Kayser
- Department of Genetic Identification, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
15
|
Si J, Chen L, Yu C, Guo Y, Sun D, Pang Y, Millwood IY, Walters RG, Yang L, Chen Y, Du H, Feng S, Yang X, Avery D, Chen J, Chen Z, Liang L, Li L, Lv J. Healthy lifestyle, DNA methylation age acceleration, and incident risk of coronary heart disease. Clin Epigenetics 2023; 15:52. [PMID: 36978155 PMCID: PMC10045869 DOI: 10.1186/s13148-023-01464-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/12/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND DNA methylation clocks emerged as a tool to determine biological aging and have been related to mortality and age-related diseases. Little is known about the association of DNA methylation age (DNAm age) with coronary heart disease (CHD), especially in the Asian population. RESULTS Methylation level of baseline blood leukocyte DNA was measured by Infinium Methylation EPIC BeadChip for 491 incident CHD cases and 489 controls in the prospective China Kadoorie Biobank. We calculated the methylation age using a prediction model developed among Chinese. The correlation between chronological age and DNAm age was 0.90. DNA methylation age acceleration (Δage) was defined as the residual of regressing DNA methylation age on the chronological age. After adjustment for multiple risk factors of CHD and cell type proportion, compared with participants in the bottom quartile of Δage, the OR (95% CI) for CHD was 1.84 (1.17, 2.89) for participants in the top quartile. One SD increment in Δage was associated with 30% increased risk of CHD (OR = 1.30; 95% CI 1.09, 1.56; Ptrend = 0.003). The average number of cigarette equivalents consumed per day and waist-to-hip ratio were positively associated with Δage; red meat consumption was negatively associated with Δage, characterized by accelerated aging in those who never or rarely consumed red meat (all P < 0.05). Further mediation analysis revealed that 10%, 5% and 18% of the CHD risk related to smoking, waist-to-hip ratio and never or rarely red meat consumption was mediated through methylation aging, respectively (all P for mediation effect < 0.05). CONCLUSIONS We first identified the association between DNAm age acceleration and incident CHD in the Asian population, and provided evidence that unfavorable lifestyle-induced epigenetic aging may play an important part in the underlying pathway to CHD.
Collapse
Affiliation(s)
- Jiahui Si
- Institute of Medical Technology, Health Science Center of Peking University, Beijing, China
- National Institute of Health Data Science at Peking University, Peking University, Beijing, China
| | - Lu Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China
- Center for Public Health and Epidemic Preparedness & Response, Peking University, 38 Xueyuan Road, Beijing, 100191, China
| | - Yu Guo
- Fuwai Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Dianjianyi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China
- Center for Public Health and Epidemic Preparedness & Response, Peking University, 38 Xueyuan Road, Beijing, 100191, China
| | - Yuanjie Pang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China
- Center for Public Health and Epidemic Preparedness & Response, Peking University, 38 Xueyuan Road, Beijing, 100191, China
| | - Iona Y Millwood
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Robin G Walters
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ling Yang
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yiping Chen
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Huaidong Du
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Shixian Feng
- NCDs Prevention and Control Department, Henan CDC, Zhengzhou, Henan, China
| | - Xiaoming Yang
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Daniel Avery
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Junshi Chen
- China National Center for Food Safety Risk Assessment, Beijing, China
| | - Zhengming Chen
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China.
- Center for Public Health and Epidemic Preparedness & Response, Peking University, 38 Xueyuan Road, Beijing, 100191, China.
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, China.
- Center for Public Health and Epidemic Preparedness & Response, Peking University, 38 Xueyuan Road, Beijing, 100191, China.
| |
Collapse
|
16
|
Ukey S, Jain A, Dwivedi S, Vishnoi JR, Chugh A, Purohit P, Pareek P, Elhence P, Misra S, Sharma P. Global and promoter specific hypermethylation of tumor suppressor genes P16, SOCS1, and SHP1 in oral squamous cell carcinoma and oral submucous fibrosis. J Cancer Res Ther 2023; 19:S551-S559. [PMID: 38384018 DOI: 10.4103/jcrt.jcrt_689_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/04/2022] [Indexed: 02/23/2024]
Abstract
ABSTRACTS Aberrant methylation pattern leads to altered gene expression, that is, involved in the transformation of various cancers, including oral squamous cell carcinoma (OSCC). In the present study, an attempt has been made to examine the association of global and promoter-specific methylation of tumor suppressor genes in patients with OSCC and oral submucous fibrosis (OSMF). Promoter-specific methylation of tumor suppressor genes P16, SOCS1, and SHP1 had been studied earlier for their aberrant methylation patterns in other cancers; however, these studies were mainly conducted in-vitro or in animal models, and as such, only a few studies are available on human samples. In the present study evaluation of promoter-specific methylation of genes P16, SOCS1, and SHP1 in 76 patients' blood and tissue samples was done and compared with methylation of 35 healthy control samples using qPCR. Further, these samples were analyzed for global methylation patterns using ELISA. The results have shown a significant decreasing trend of promoter methylation (OSCC > OSMF > Controls); the methylation indices (MI) were significantly higher in OSCC than in the controls. The median MI of three genes for OSCC were P16MI (0.96), SHP1MI (0.79), and SOCS1 (0.80). Similarly, median MIs for OSMF were P16MI (0.18), SHP1 MI (0.19), and SOCS1 MI (0.5) against controls with MI (0) for each of the three genes. The global methylation %mC values were 1.9, 0.5, and 0.1, respectively. The values of MI and %mC were found to correlate with various risk factors such as tobacco, smoking, and alcohol consumption, which are positively involved in OSMF pathogenesis followed by oral cancer progression. Further, the methylation trend in tissue was reflected in blood samples, proving a window for methylation load to be used as a lesser invasive biomarker. The sensitivity and specificity of methylation load were also found reasonable. Therefore, the current study suggests that there may be a role of global and promoter-specific methylation load in the transition of OSMF to OSCC.
Collapse
Affiliation(s)
- Shweta Ukey
- All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Ankit Jain
- All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Shailendra Dwivedi
- All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India
| | | | - Ankita Chugh
- All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Purvi Purohit
- All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Puneet Pareek
- All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Poonam Elhence
- All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Sanjeev Misra
- All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Praveen Sharma
- All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| |
Collapse
|
17
|
Cabrera-Mendoza B, Stertz L, Najera K, Selvaraj S, Teixeira AL, Meyer TD, Fries GR, Walss-Bass C. Within subject cross-tissue analyzes of epigenetic clocks in substance use disorder postmortem brain and blood. Am J Med Genet B Neuropsychiatr Genet 2023; 192:13-27. [PMID: 36056652 PMCID: PMC9742183 DOI: 10.1002/ajmg.b.32920] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/22/2022] [Accepted: 08/16/2022] [Indexed: 12/14/2022]
Abstract
There is a possible accelerated biological aging in patients with substance use disorders (SUD). The evaluation of epigenetic clocks, which are accurate estimators of biological aging based on DNA methylation changes, has been limited to blood tissue in patients with SUD. Consequently, the impact of biological aging in the brain of individuals with SUD remains unknown. In this study, we evaluated multiple epigenetic clocks (DNAmAge, DNAmAgeHannum, DNAmAgeSkinBlood, DNAmPhenoAge, DNAmGrimAge, and DNAmTL) in individuals with SUD (n = 42), including alcohol (n = 10), opioid (n = 19), and stimulant use disorder (n = 13), and controls (n = 10) in postmortem brain (prefrontal cortex) and blood tissue obtained from the same individuals. We found a higher DNAmPhenoAge (β = 0.191, p-value = 0.0104) and a nominally lower DNAmTL (β = -0.149, p-value = 0.0603) in blood from individuals with SUD compared to controls. SUD subgroup analysis showed a nominally lower brain DNAmTL in subjects with alcohol use disorder, compared to stimulant use disorder and controls (β = 0.0150, p-value = 0.087). Cross-tissue analyzes indicated a lower blood DNAmTL and a higher blood DNAmAge compared to their respective brain values in the SUD group. This study highlights the relevance of tissue specificity in biological aging studies and suggests that peripheral measures of epigenetic clocks in SUD may depend on the specific type of drug used.
Collapse
Affiliation(s)
- Brenda Cabrera-Mendoza
- PECEM, Faculty of Medicine, Universidad Nacional
Autónoma de México, Mexico City, 04510, Mexico
| | - Laura Stertz
- Louis A. Faillace, MD, Department of Psychiatry and
Behavioral Sciences, McGovern Medical School, University of Texas Health Science
Center at Houston, Houston, TX, 77054, USA
| | - Katherine Najera
- Louis A. Faillace, MD, Department of Psychiatry and
Behavioral Sciences, McGovern Medical School, University of Texas Health Science
Center at Houston, Houston, TX, 77054, USA
| | - Sudhakar Selvaraj
- Louis A. Faillace, MD, Department of Psychiatry and
Behavioral Sciences, McGovern Medical School, University of Texas Health Science
Center at Houston, Houston, TX, 77054, USA
| | - Antonio L. Teixeira
- Louis A. Faillace, MD, Department of Psychiatry and
Behavioral Sciences, McGovern Medical School, University of Texas Health Science
Center at Houston, Houston, TX, 77054, USA
| | - Thomas D. Meyer
- Louis A. Faillace, MD, Department of Psychiatry and
Behavioral Sciences, McGovern Medical School, University of Texas Health Science
Center at Houston, Houston, TX, 77054, USA
| | - Gabriel R. Fries
- Louis A. Faillace, MD, Department of Psychiatry and
Behavioral Sciences, McGovern Medical School, University of Texas Health Science
Center at Houston, Houston, TX, 77054, USA
- Center for Precision Health, School of Biomedical
Informatics, University of Texas Health Science Center at Houston, Houston, TX,
77054, USA
| | - Consuelo Walss-Bass
- Louis A. Faillace, MD, Department of Psychiatry and
Behavioral Sciences, McGovern Medical School, University of Texas Health Science
Center at Houston, Houston, TX, 77054, USA
| |
Collapse
|
18
|
Liu C, Wang Z, Hui Q, Goldberg J, Smith NL, Shah AJ, Murrah N, Shallenberger L, Diggers E, Bremner JD, Sun YV, Vaccarino V. Association between depression and epigenetic age acceleration: A co-twin control study. Depress Anxiety 2022; 39:741-750. [PMID: 35758529 PMCID: PMC9729366 DOI: 10.1002/da.23279] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 05/26/2022] [Accepted: 06/18/2022] [Indexed: 01/27/2023] Open
Abstract
INTRODUCTION Prior studies have shown inconsistent findings of an association between depression and epigenetic aging. DNA methylation (DNAm) age acceleration can measure biological aging. We adopted a robust co-twin control study design to examine whether depression is associated with DNAm age acceleration after accounting for the potential confounding influences of genetics and family environment. METHODS We analyzed data on a sub-cohort of the Vietnam Era Twin Registry. A total of 291 twins participated at baseline and 177 at follow-up visit after a mean of 11.7 years, with 111 participants having DNA samples for both time points. Depression was measured using the Beck Depression Inventory II (BDI-II). Six measures of DNAm age acceleration were computed at each time point, including Horvath's DNAm age acceleration (HorvathAA), intrinsic epigenetic age acceleration (IEAA), Hannum's DNAm age acceleration (HannumAA), extrinsic epigenetic age acceleration (EEAA), GrimAge acceleration (GrimAA), and PhenoAge acceleration (PhenoAA). Mixed-effects modeling was used to assess the within-pair association between depression and DNAm age acceleration. RESULTS At baseline, a 10-unit higher BDI-II total score was associated with HannumAA (0.73 years, 95% confidence interval [CI] 0.13-1.33, p = .019) and EEAA (0.94 years, 95% CI 0.22-1.66, p = .012). At follow-up, 10-unit higher BDI-II score was associated with PhenoAA (1.32 years, 95% CI 0.18-2.47, p = .027). CONCLUSION We identified that depression is associated with higher levels of DNAm age acceleration. Further investigation is warranted to better understand the underlying mechanisms for the potential causal relationship between depression and accelerated aging.
Collapse
Affiliation(s)
- Chang Liu
- Department of Epidemiology, Emory University Rollins School of Public Health, 1518 Clifton Road NE, Atlanta, GA
| | - Zeyuan Wang
- Department of Epidemiology, Emory University Rollins School of Public Health, 1518 Clifton Road NE, Atlanta, GA
| | - Qin Hui
- Department of Epidemiology, Emory University Rollins School of Public Health, 1518 Clifton Road NE, Atlanta, GA
| | - Jack Goldberg
- Vietnam Era Twin Registry, Seattle Epidemiologic Research and Information Center, Cooperative Studies Program, Office of Research and Development, Department of Veterans Affairs, Seattle, WA, USA
| | - Nicholas L. Smith
- Vietnam Era Twin Registry, Seattle Epidemiologic Research and Information Center, Cooperative Studies Program, Office of Research and Development, Department of Veterans Affairs, Seattle, WA, USA
| | - Amit J. Shah
- Department of Epidemiology, Emory University Rollins School of Public Health, 1518 Clifton Road NE, Atlanta, GA
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Atlanta VA Health Care System, 1670 Clairmont Road, Decatur, GA 30033, USA
| | - Nancy Murrah
- Department of Epidemiology, Emory University Rollins School of Public Health, 1518 Clifton Road NE, Atlanta, GA
| | - Lucy Shallenberger
- Department of Epidemiology, Emory University Rollins School of Public Health, 1518 Clifton Road NE, Atlanta, GA
| | - Emily Diggers
- Department of Epidemiology, Emory University Rollins School of Public Health, 1518 Clifton Road NE, Atlanta, GA
| | - J. Douglas Bremner
- Atlanta VA Health Care System, 1670 Clairmont Road, Decatur, GA 30033, USA
- Departments of Psychiatry and Behavioral Sciences and Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA
| | - Yan V. Sun
- Department of Epidemiology, Emory University Rollins School of Public Health, 1518 Clifton Road NE, Atlanta, GA
- Atlanta VA Health Care System, 1670 Clairmont Road, Decatur, GA 30033, USA
| | - Viola Vaccarino
- Department of Epidemiology, Emory University Rollins School of Public Health, 1518 Clifton Road NE, Atlanta, GA
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
19
|
Fuggle NR, Laskou F, Harvey NC, Dennison EM. A review of epigenetics and its association with ageing of muscle and bone. Maturitas 2022; 165:12-17. [PMID: 35841774 DOI: 10.1016/j.maturitas.2022.06.014] [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: 04/30/2022] [Revised: 06/21/2022] [Accepted: 06/30/2022] [Indexed: 10/31/2022]
Abstract
Ageing is defined as the 'increasing frailty of an organism with time that reduces the ability of that organism to deal with stress'. It has been suggested that epigenetics may underlie the observation that some individuals appear to age faster than others. Epigenetics is the study of changes which occur in an organism due to changes in expression of the genetic code rather than changes to the genetic code itself; that is, epigenetic mechanisms impact upon the function of DNA without changing the DNA sequence. It is important to recognise that epigenetic changes, in contrast to genetic changes, can vary according to different cell types and therefore can demonstrate significant tissue-specificity. There are different types of epigenetic mechanisms: histone modification, non-coding RNAs and DNA methylation. Epigenetic clocks have been developed using statistical techniques to identify the optimal combination of CpG sites (from methylation arrays) to correlate with chronological age. This review considers how epigenetic factors may affect rates of ageing of muscle and bone and provides an overview of current understanding in this area. We discuss studies using first-generation epigenetic clocks, as well as the second-generation iterations, which appear to show stronger associations with the ageing muscle phenotype. We also review epigenome-wide association studies that have been performed in various tissues examining relationships with osteoporosis and fracture. It is hoped that an understanding of this area will lead to interventions that might prevent or reduce rates of musculoskeletal ageing in later life.
Collapse
Affiliation(s)
- N R Fuggle
- MRC Lifecourse Epidemiology Centre, University of Southampton, SO16 6YD, United Kingdom of Great Britain and Northern Ireland
| | - F Laskou
- MRC Lifecourse Epidemiology Centre, University of Southampton, SO16 6YD, United Kingdom of Great Britain and Northern Ireland
| | - N C Harvey
- MRC Lifecourse Epidemiology Centre, University of Southampton, SO16 6YD, United Kingdom of Great Britain and Northern Ireland
| | - E M Dennison
- MRC Lifecourse Epidemiology Centre, University of Southampton, SO16 6YD, United Kingdom of Great Britain and Northern Ireland.
| |
Collapse
|
20
|
Schmitz LL, Zhao W, Ratliff SM, Goodwin J, Miao J, Lu Q, Guo X, Taylor KD, Ding J, Liu Y, Levine M, Smith JA. The Socioeconomic Gradient in Epigenetic Ageing Clocks: Evidence from the Multi-Ethnic Study of Atherosclerosis and the Health and Retirement Study. Epigenetics 2022; 17:589-611. [PMID: 34227900 PMCID: PMC9235889 DOI: 10.1080/15592294.2021.1939479] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/02/2021] [Indexed: 12/25/2022] Open
Abstract
Epigenetic clocks have been widely used to predict disease risk in multiple tissues or cells. Their success as a measure of biological ageing has prompted research on the connection between epigenetic pathways of ageing and the socioeconomic gradient in health and mortality. However, studies examining social correlates of epigenetic ageing have yielded inconsistent results. We conducted a comprehensive, comparative analysis of associations between various dimensions of socioeconomic status (SES) (education, income, wealth, occupation, neighbourhood environment, and childhood SES) and eight epigenetic clocks in two well-powered US ageing studies: The Multi-Ethnic Study of Atherosclerosis (MESA) (n = 1,211) and the Health and Retirement Study (HRS) (n = 4,018). In both studies, we found robust associations between SES measures in adulthood and the GrimAge and DunedinPoAm clocks (Bonferroni-corrected p-value < 0.01). In the HRS, significant associations with the Levine and Yang clocks were also evident. These associations were only partially mediated by smoking, alcohol consumption, and obesity, which suggests that differences in health behaviours alone cannot explain the SES gradient in epigenetic ageing in older adults. Further analyses revealed concurrent associations between polygenic risk for accelerated intrinsic epigenetic ageing, SES, and the Levine clock, indicating that genetic risk and social disadvantage may contribute additively to faster biological aging.
Collapse
Affiliation(s)
- Lauren L. Schmitz
- Robert M. La Follette School of Public Affairs, University of Wisconsin-Madison, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, USA
| | - Scott M. Ratliff
- Department of Epidemiology, School of Public Health, University of Michigan, USA
| | - Julia Goodwin
- Department of Sociology, University of Wisconsin-Madison, USA
| | - Jiacheng Miao
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, USA
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, USA
- Department of Statistics, University of Wisconsin-Madison, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, USA
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, USA
| | - Jingzhong Ding
- Gerontology and Geriatric Medicine, School of Medicine, Wake Forest University, USA
| | - Yongmei Liu
- Department of Medicine, School of Medicine, Duke University, USA
| | - Morgan Levine
- Department of Pathology, School of Medicine, Yale University, USA
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, USA
- Survey Research Center, Institute for Social Research, University of Michigan, USA
| |
Collapse
|
21
|
Klopack ET, Carroll JE, Cole SW, Seeman TE, Crimmins EM. Lifetime exposure to smoking, epigenetic aging, and morbidity and mortality in older adults. Clin Epigenetics 2022; 14:72. [PMID: 35643537 PMCID: PMC9148451 DOI: 10.1186/s13148-022-01286-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/09/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cigarette smoke is a major public health concern. Epigenetic aging may be an important pathway by which exposure to cigarette smoke affects health. However, little is known about how exposure to smoke at different life stages affects epigenetic aging, especially in older adults. This study examines how three epigenetic aging measures (GrimAge, PhenoAge, and DunedinPoAm38) are associated with parental smoking, smoking in youth, and smoking in adulthood, and whether these epigenetic aging measures mediate the link between smoke exposure and morbidity and mortality. This study utilizes data from the Health and Retirement Study (HRS) Venous Blood Study (VBS), a nationally representative sample of US adults over 50 years old collected in 2016. 2978 participants with data on exposure to smoking, morbidity, and mortality were included. RESULTS GrimAge is significantly increased by having two smoking parents, smoking in youth, and cigarette pack years in adulthood. PhenoAge and DunedinPoAm38 are associated with pack years. All three mediate some of the effect of pack years on cancer, high blood pressure, heart disease, and mortality and GrimAge and DunedinPoAm38 mediate this association on lung disease. CONCLUSIONS Results suggest epigenetic aging is one biological mechanism linking lifetime exposure to smoking with development of disease and earlier death in later life. Interventions aimed at reducing smoking in adulthood may be effective at weakening this association.
Collapse
Affiliation(s)
- Eric T Klopack
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave., Los Angeles, CA, 90089, USA.
| | - Judith E Carroll
- Cousins Center for Psychoneuroimmunology, Department of Psychiatry & Biobehavioral Sciences, Jane & Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Steve W Cole
- Cousins Center for Psychoneuroimmunology, Jane & Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Teresa E Seeman
- Division of Geriatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Eileen M Crimmins
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave., Los Angeles, CA, 90089, USA
| |
Collapse
|
22
|
Dada O, Jeremian R, Dai N, Strauss J, Zai C, Graff A, De Luca V. Investigation of accelerated epigenetic aging in individuals experiencing suicidal ideation. Schizophr Res 2022; 243:307-309. [PMID: 33041163 DOI: 10.1016/j.schres.2020.09.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 09/17/2020] [Accepted: 09/26/2020] [Indexed: 10/23/2022]
Affiliation(s)
| | - Richie Jeremian
- CAMH, Department of Psychiatry, University of Toronto, Canada
| | - Nasia Dai
- CAMH, Department of Psychiatry, University of Toronto, Canada
| | - John Strauss
- CAMH, Department of Psychiatry, University of Toronto, Canada
| | - Clement Zai
- CAMH, Department of Psychiatry, University of Toronto, Canada
| | - Ariel Graff
- CAMH, Department of Psychiatry, University of Toronto, Canada
| | | |
Collapse
|
23
|
Lopuszanska-Dawid M, Kołodziej H, Lipowicz A, Szklarska A. Age, Education, and Stress Affect Ageing Males' Symptoms More than Lifestyle Does: The Wroclaw Male Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19095044. [PMID: 35564437 PMCID: PMC9105921 DOI: 10.3390/ijerph19095044] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 12/13/2022]
Abstract
An increasing number of subjects are affected by health problems related to the advanced involutional processes. It is extremely important to identify the determinants of the rate of occurrence of physiological, psychological, and social manifestations of aging. The aim was to determine how factors such as lifestyle, level of education, or severity of stressful life events indicate the appearance of aging symptoms in adult men. The material consisted of data of ethnically homogeneous group of 355 men (32−87 years), invited to the study as a part of the Wroclaw Male Study research project. The analyzed features included (1) socioeconomic status: age, educational level, marital status, and having children; (2) elements of lifestyle: alcohol drinking, cigarette smoking, and physical activity; (3) major and most important stressful life events—the Social Readjustment Rating Scale; (4) symptoms related to male aging—the Aging Males’ Symptoms. The backward stepwise regression models, the Kruskal−Wallis test, and multiple comparisons of mean ranks were used. Noncentrality parameter δ (delta), two-tailed critical values of the test, and test power with α = 0.05 were calculated. Among the analyzed variables, age was most strongly associated with the intensity of almost all groups of andropausal symptoms in men (p = 0.0001), followed by the level of education (p = 0.0001) and the intensity of stressful life events (p = 0.0108). Selected lifestyle elements turned out to be much less important (p > 0.01). Preventive actions aimed at slowing down the intensification of involutional processes, including teaching strategies for coping with stressful life events, should be implemented in groups of men with specific risk factors from an early age.
Collapse
Affiliation(s)
- Monika Lopuszanska-Dawid
- Department of Human Biology, Faculty of Physical Education, Józef Piłsudski University of Physical Education in Warsaw, Marymoncka 34, 00-968 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-834-04-31
| | - Halina Kołodziej
- Department of Anthropology, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, C. K. Norwida 25, 50-375 Wroclaw, Poland; (H.K.); (A.L.)
| | - Anna Lipowicz
- Department of Anthropology, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, C. K. Norwida 25, 50-375 Wroclaw, Poland; (H.K.); (A.L.)
| | - Alicja Szklarska
- Polish Academy of Sciences, Palace of Culture and Science, Defilad Square 1, 00-901 Warsaw, Poland;
| |
Collapse
|
24
|
Hibernation slows epigenetic ageing in yellow-bellied marmots. Nat Ecol Evol 2022; 6:418-426. [PMID: 35256811 PMCID: PMC8986532 DOI: 10.1038/s41559-022-01679-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 01/20/2022] [Indexed: 01/02/2023]
Abstract
Species that hibernate generally live longer than would be expected based solely on their body size. Hibernation is characterized by long periods of metabolic suppression (torpor) interspersed by short periods of increased metabolism (arousal). The torpor–arousal cycles occur multiple times during hibernation, and it has been suggested that processes controlling the transition between torpor and arousal states cause ageing suppression. Metabolic rate is also a known correlate of longevity; we thus proposed the ‘hibernation–ageing hypothesis’ whereby ageing is suspended during hibernation. We tested this hypothesis in a well-studied population of yellow-bellied marmots (Marmota flaviventer), which spend 7–8 months per year hibernating. We used two approaches to estimate epigenetic age: the epigenetic clock and the epigenetic pacemaker. Variation in epigenetic age of 149 samples collected throughout the life of 73 females was modelled using generalized additive mixed models (GAMM), where season (cyclic cubic spline) and chronological age (cubic spline) were fixed effects. As expected, the GAMM using epigenetic ages calculated from the epigenetic pacemaker was better able to detect nonlinear patterns in epigenetic ageing over time. We observed a logarithmic curve of epigenetic age with time, where the epigenetic age increased at a higher rate until females reached sexual maturity (two years old). With respect to circannual patterns, the epigenetic age increased during the active season and essentially stalled during the hibernation period. Taken together, our results are consistent with the hibernation–ageing hypothesis and may explain the enhanced longevity in hibernators. Species that hibernate generally have longer lifespans than expected based on their body size. The authors show epigenetic ageing patterns from a natural population of hibernating yellow-bellied marmots consistent with the hypothesis that ageing is suspended during hibernation.
Collapse
|
25
|
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: 24] [Impact Index Per Article: 12.0] [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
|
26
|
Abstract
As the most phenotypically diverse mammalian species that shares human environments and access to sophisticated healthcare, domestic dogs have unique potential to inform our understanding of the determinants of aging. Here we outline key concepts in the study of aging and illustrate the value of research with dogs, which can improve dog health and support translational discoveries. We consider similarities and differences in aging and age-related diseases in dogs and humans and summarize key advances in our understanding of genetic and environmental risk factors for morbidity and mortality in dogs. We address health outcomes ranging from cancer to cognitive function and highlight emerging research opportunities from large-scale cohort studies in companion dogs. We conclude that studying aging in dogs could overcome many limitations of laboratory models, most notably, the ability to assess how aging-associated pathways influence aging in real-world environments similar to those experienced by humans.
Collapse
Affiliation(s)
- Audrey Ruple
- Department of Population Health Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA;
| | - Evan MacLean
- School of Anthropology and College of Veterinary Medicine, University of Arizona, Tucson, Arizona, USA;
| | - Noah Snyder-Mackler
- School of Life Sciences, Center for Evolution and Medicine, and School for Human Evolution and Social Change, Arizona State University, Tempe, Arizona, USA;
| | - Kate E. Creevy
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Daniel Promislow
- Department of Laboratory Medicine & Pathology and Department of Biology, University of Washington School of Medicine, Seattle, Washington, USA;
| |
Collapse
|
27
|
Soda K. Overview of Polyamines as Nutrients for Human Healthy Long Life and Effect of Increased Polyamine Intake on DNA Methylation. Cells 2022; 11:cells11010164. [PMID: 35011727 PMCID: PMC8750749 DOI: 10.3390/cells11010164] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 02/04/2023] Open
Abstract
Polyamines, spermidine and spermine, are synthesized in every living cell and are therefore contained in foods, especially in those that are thought to contribute to health and longevity. They have many physiological activities similar to those of antioxidant and anti-inflammatory substances such as polyphenols. These include antioxidant and anti-inflammatory properties, cell and gene protection, and autophagy activation. We have first reported that increased polyamine intake (spermidine much more so than spermine) over a long period increased blood spermine levels and inhibited aging-associated pathologies and pro-inflammatory status in humans and mice and extended life span of mice. However, it is unlikely that the life-extending effect of polyamines is exerted by the same bioactivity as polyphenols because most studies using polyphenols and antioxidants have failed to demonstrate their life-extending effects. Recent investigations revealed that aging-associated pathologies and lifespan are closely associated with DNA methylation, a regulatory mechanism of gene expression. There is a close relationship between polyamine metabolism and DNA methylation. We have shown that the changes in polyamine metabolism affect the concentrations of substances and enzyme activities involved in DNA methylation. I consider that the increased capability of regulation of DNA methylation by spermine is a key of healthy long life of humans.
Collapse
Affiliation(s)
- Kuniyasu Soda
- Department Cardiovascular Institute for Medical Research, Saitama Medical Center, Jichi Medical University, 1-847, Amanuma, Saitama-City 330-0834, Saitama, Japan
| |
Collapse
|
28
|
Ohmomo H, Harada S, Komaki S, Ono K, Sutoh Y, Otomo R, Umekage S, Hachiya T, Katanoda K, Takebayashi T, Shimizu A. DNA Methylation Abnormalities and Altered Whole Transcriptome Profiles after Switching from Combustible Tobacco Smoking to Heated Tobacco Products. Cancer Epidemiol Biomarkers Prev 2022; 31:269-279. [PMID: 34728466 PMCID: PMC9398167 DOI: 10.1158/1055-9965.epi-21-0444] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/29/2021] [Accepted: 10/18/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The use of heated tobacco products (HTP) has increased exponentially in Japan since 2016; however, their effects on health remain a major concern. METHODS Tsuruoka Metabolome Cohort Study participants (n = 11,002) were grouped on the basis of their smoking habits as never smokers (NS), past smokers (PS), combustible tobacco smokers (CS), and HTP users for <2 years. Peripheral blood mononuclear cells were collected from 52 participants per group matched to HTP users using propensity scores, and DNA and RNA were purified from the samples. DNA methylation (DNAm) analysis of the 17 smoking-associated DNAm biomarker genes (such as AHRR, F2RL3, LRRN3, and GPR15), as well as whole transcriptome analysis, was performed. RESULTS Ten of the 17 genes were significantly hypomethylated in CS and HTP users compared with NS, among which AHRR, F2RL3, and RARA showed intermediate characteristics between CS and NS; nonetheless, AHRR expression was significantly higher in CS than in the other three groups. Conversely, LRRN3 and GPR15 were more hypomethylated in HTP users than in NS, and GPR15 expression was markedly upregulated in all the groups when compared with that in NS. CONCLUSIONS HTP users (switched from CS <2 years) display abnormal DNAm and transcriptome profiles, albeit to a lesser extent than the CS. However, because the molecular genetic effects of long-term HTP use are still unknown, long-term molecular epidemiologic studies are needed. IMPACT This study provides new insights into the molecular genetic effects on DNAm and transcriptome profiles in HTP users who switched from CS.
Collapse
Affiliation(s)
- Hideki Ohmomo
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Yahaba, Shiwa, Iwate, Japan
| | - Sei Harada
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Shohei Komaki
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Yahaba, Shiwa, Iwate, Japan
| | - Kanako Ono
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Yahaba, Shiwa, Iwate, Japan
| | - Yoichi Sutoh
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Yahaba, Shiwa, Iwate, Japan
| | - Ryo Otomo
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Yahaba, Shiwa, Iwate, Japan
| | - So Umekage
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Yahaba, Shiwa, Iwate, Japan
| | - Tsuyoshi Hachiya
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Yahaba, Shiwa, Iwate, Japan
| | - Kota Katanoda
- Division of Cancer Statistics Integration, National Cancer Center Research Institute, Chuo, Tokyo, Japan
| | - Toru Takebayashi
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Atsushi Shimizu
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Yahaba, Shiwa, Iwate, Japan.,Corresponding Author: Atsushi Shimizu, Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate 028-3694, Japan. Phone: 81-19-651-5110, ext. 5473; E-mail:
| |
Collapse
|
29
|
Chung M, Ruan M, Zhao N, Koestler DC, De Vivo I, Kelsey KT, Michaud DS. DNA methylation ageing clocks and pancreatic cancer risk: pooled analysis of three prospective nested case-control studies. Epigenetics 2021; 16:1306-1316. [PMID: 33315530 PMCID: PMC8813079 DOI: 10.1080/15592294.2020.1861401] [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: 08/06/2020] [Revised: 10/06/2020] [Accepted: 11/19/2020] [Indexed: 10/22/2022] Open
Abstract
DNA methylation (DNAm) age may reflect age-related variations in biological changes and abnormalities related to ageing. DNAm age acceleration measures have been associated with a number of cancers, but to our knowledge, have not been examined in relation to pancreatic cancer risk or survival. DNAm levels in leukocytes of prediagnostic blood samples of 393 pancreatic cancer cases and 431 matched controls, pooled from three large prospective cohort studies, were used to estimate DNAm age, epigenetic age acceleration (AA), and intrinsic epigenetic age acceleration (IEAA) metrics. Logistic regression and Cox proportional hazard regression models were used to examine the relationship between the various AA and IEAA metrics and pancreatic cancer risk and survival, respectively. The results showed that pancreatic cancer risk was significantly increased across all IEAA metrics, ranging from 83% to 95% increased risk when comparing the third and highest quartiles to the lowest quartile of IEAA. Consistent with these findings, the results from multivariate spline regression analyses showed non-linear relationships between all three IEAA metrics and pancreatic cancer risk with apparent threshold effect including two turning points at minimal and at maximal risks, respectively. There is no evidence of a significant association between pancreatic cancer survival and any of the epigenetic AA or IEAA metrics. Our results indicate DNAm age acceleration, measured in blood prior to cancer diagnosis, is associated with an increased risk of pancreatic cancer in a complex nonlinear, dose-response manner. Epigenetic IEAA metrics may be a useful addition to current methods for pancreatic cancer risk prediction.
Collapse
Affiliation(s)
- Mei Chung
- Department of Public Health & Community Medicine, Tufts University School of Medicine, Tufts University, Boston, MA, USA
| | - Mengyuan Ruan
- Department of Public Health & Community Medicine, Tufts University School of Medicine, Tufts University, Boston, MA, USA
| | - Naisi Zhao
- Department of Public Health & Community Medicine, Tufts University School of Medicine, Tufts University, Boston, MA, USA
| | - Devin C. Koestler
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Immaculata De Vivo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Karl T. Kelsey
- Department of Epidemiology, Brown University, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Dominique S. Michaud
- Department of Public Health & Community Medicine, Tufts University School of Medicine, Tufts University, Boston, MA, USA
| |
Collapse
|
30
|
Ritonja JA, Aronson KJ, Flaten L, Topouza DG, Duan QL, Durocher F, Tranmer JE, Bhatti P. Exploring the impact of night shift work on methylation of circadian genes. Epigenetics 2021; 17:1259-1268. [PMID: 34825628 DOI: 10.1080/15592294.2021.2009997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Night shift work is associated with increased breast cancer risk, but the molecular mechanisms are not well-understood. The objective of this study was to explore the relationship between night shift work parameters (current status, duration/years, and intensity) and methylation in circadian genes as a potential mechanism underlying the carcinogenic effects of night shift work. A cross-sectional study was conducted among 74 female healthcare employees (n = 38 day workers, n = 36 night shift workers). The Illumina Infinium MethylationEPIC beadchip was applied to DNA extracted from blood samples to measure methylation using a candidate gene approach at 1150 CpG loci across 22 circadian genes. Linear regression models were used to examine the association between night shift work parameters and continuous methylation measurements (β-values) for each CpG site. The false-discovery rate (q = 0.2) was used to account for multiple comparisons. Compared to day workers, current night shift workers demonstrated hypermethylation in the 5'UTR region of CSNK1E (q = 0.15). Individuals that worked night shifts for ≥10 years exhibited hypomethylation in the gene body of NR1D1 (q = 0.08) compared to those that worked <10 years. Hypermethylation in the gene body of ARNTL was also apparent in those who worked ≥3 consecutive night shifts a week (q = 0.18). These findings suggest that night shift work is associated with differential methylation in core circadian genes, including CSNK1E, NR1D1 and ARNTL. Future, larger-scale studies with long-term follow-up and detailed night shift work assessment are needed to confirm and expand on these findings.
Collapse
Affiliation(s)
- Jennifer A Ritonja
- Department of Public Health Sciences, Queen's University, Kingston, Canada
| | - Kristan J Aronson
- Department of Public Health Sciences, Queen's University, Kingston, Canada.,Division of Cancer Care and Epidemiology, Cancer Research Institute, Queen's University, Kingston, Canada
| | - Lisa Flaten
- Department of Public Health Sciences, Queen's University, Kingston, Canada
| | - Danai G Topouza
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Qing Ling Duan
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada.,School of Computing, Queen's University, Kingston, Canada
| | - Francine Durocher
- Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Kingston, Canada.,Centre de Recherche Sur Le Cancer, Centre de Recherche Du Chu de Québec-Université Laval, Quebec, Canada
| | - Joan E Tranmer
- Department of Public Health Sciences, Queen's University, Kingston, Canada.,The School of Nursing is the department, School of Nursing, Queen's University, Kingston, Canada
| | | |
Collapse
|
31
|
de Prado-Bert P, Ruiz-Arenas C, Vives-Usano M, Andrusaityte S, Cadiou S, Carracedo Á, Casas M, Chatzi L, Dadvand P, González JR, Grazuleviciene R, Gutzkow KB, Haug LS, Hernandez-Ferrer C, Keun HC, Lepeule J, Maitre L, McEachan R, Nieuwenhuijsen MJ, Pelegrí D, Robinson O, Slama R, Vafeiadi M, Sunyer J, Vrijheid M, Bustamante M. The early-life exposome and epigenetic age acceleration in children. ENVIRONMENT INTERNATIONAL 2021; 155:106683. [PMID: 34144479 DOI: 10.1016/j.envint.2021.106683] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/01/2021] [Accepted: 06/01/2021] [Indexed: 06/12/2023]
Abstract
The early-life exposome influences future health and accelerated biological aging has been proposed as one of the underlying biological mechanisms. We investigated the association between more than 100 exposures assessed during pregnancy and in childhood (including indoor and outdoor air pollutants, built environment, green environments, tobacco smoking, lifestyle exposures, and biomarkers of chemical pollutants), and epigenetic age acceleration in 1,173 children aged 7 years old from the Human Early-Life Exposome project. Age acceleration was calculated based on Horvath's Skin and Blood clock using child blood DNA methylation measured by Infinium HumanMethylation450 BeadChips. We performed an exposure-wide association study between prenatal and childhood exposome and age acceleration. Maternal tobacco smoking during pregnancy was nominally associated with increased age acceleration. For childhood exposures, indoor particulate matter absorbance (PMabs) and parental smoking were nominally associated with an increase in age acceleration. Exposure to the organic pesticide dimethyl dithiophosphate and the persistent pollutant polychlorinated biphenyl-138 (inversely associated with child body mass index) were protective for age acceleration. None of the associations remained significant after multiple-testing correction. Pregnancy and childhood exposure to tobacco smoke and childhood exposure to indoor PMabs may accelerate epigenetic aging from an early age.
Collapse
Affiliation(s)
- Paula de Prado-Bert
- ISGlobal, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Carlos Ruiz-Arenas
- ISGlobal, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Marta Vives-Usano
- ISGlobal, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Sandra Andrusaityte
- Department of Environmental Sciences, Vytautas Magnus University, K. Donelaicio Street 58, 44248 Kaunas, Lithuania
| | - Solène Cadiou
- University Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, IAB, 38000 Grenoble, France
| | - Ángel Carracedo
- Grupo de Medicina Xenómica, Fundación Pública Galega de Medicina Xenómica, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), SERGAS, Rúa Choupana s/n, 15706 Santiago de Compostela, Spain; Centro de Investigación en Red de Enfermedades Raras (CIBERER) y Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII), Universidade de Santiago de Compostela, Praza do Obradoiro s/n, 15782 Santiago de Compostela, Spain
| | - Maribel Casas
- ISGlobal, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Leda Chatzi
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, USA; Department of Social Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Payam Dadvand
- ISGlobal, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Juan R González
- ISGlobal, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Regina Grazuleviciene
- Department of Environmental Sciences, Vytautas Magnus University, K. Donelaicio Street 58, 44248 Kaunas, Lithuania
| | - Kristine B Gutzkow
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Line S Haug
- Department of Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Carles Hernandez-Ferrer
- ISGlobal, Dr. Aiguader 88, 08003 Barcelona, Spain; CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Baldiri Reixac 4, Barcelona 08028, Spain
| | - Hector C Keun
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Johanna Lepeule
- University Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, IAB, 38000 Grenoble, France
| | - Léa Maitre
- ISGlobal, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Rosie McEachan
- Bradford Institute for Health Research, Bradford Royal Infirmary, Bradford, UK
| | - Mark J Nieuwenhuijsen
- ISGlobal, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Dolors Pelegrí
- ISGlobal, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Oliver Robinson
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, St Marys Hospital Campus, London W21PG, UK
| | - Rémy Slama
- University Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, IAB, 38000 Grenoble, France
| | - Marina Vafeiadi
- Department of Social Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Jordi Sunyer
- ISGlobal, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Martine Vrijheid
- ISGlobal, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Mariona Bustamante
- ISGlobal, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain.
| |
Collapse
|
32
|
Piniewska-Róg D, Heidegger A, Pośpiech E, Xavier C, Pisarek A, Jarosz A, Woźniak A, Wojtas M, Phillips C, Kayser M, Parson W, Branicki W. Impact of excessive alcohol abuse on age prediction using the VISAGE enhanced tool for epigenetic age estimation in blood. Int J Legal Med 2021; 135:2209-2219. [PMID: 34405265 PMCID: PMC8523459 DOI: 10.1007/s00414-021-02665-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/06/2021] [Indexed: 12/13/2022]
Abstract
DNA methylation-based clocks provide the most accurate age estimates with practical implications for clinical and forensic genetics. However, the effects of external factors that may influence the estimates are poorly studied. Here, we evaluated the effect of alcohol consumption on epigenetic age prediction in a cohort of extreme alcohol abusers. Blood samples from deceased alcohol abusers and age- and sex-matched controls were analyzed using the VISAGE enhanced tool for age prediction from somatic tissues that enables examination of 44 CpGs within eight age markers. Significantly altered DNA methylation was recorded for alcohol abusers in MIR29B2CHG. This resulted in a mean predicted age of 1.4 years higher compared to the controls and this trend increased in older individuals. The association of alcohol abuse with epigenetic age acceleration, as determined by the prediction analysis performed based on MIR29B2CHG, was small but significant (β = 0.190; P-value = 0.007). However, the observed alteration in DNA methylation of MIR29B2CHG had a non-significant effect on age estimation with the VISAGE age prediction model. The mean absolute error in the alcohol-abusing cohort was 3.1 years, compared to 3.3 years in the control group. At the same time, upregulation of MIR29B2CHG expression may have a biological function, which merits further studies.
Collapse
Affiliation(s)
- Danuta Piniewska-Róg
- Jagiellonian University Medical College, Faculty of Medicine, Department of Forensic Medicine, Grzegórzecka 16, 31-531, Krakow, Poland
| | - Antonia Heidegger
- Institute of Legal Medicine, Medical University of Innsbruck, Muellerstrasse 44, 6020, Innsbruck, Austria
| | - Ewelina Pośpiech
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-348, Krakow, Poland
| | - Catarina Xavier
- Institute of Legal Medicine, Medical University of Innsbruck, Muellerstrasse 44, 6020, Innsbruck, Austria
| | - Aleksandra Pisarek
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-348, Krakow, Poland
| | - Agata Jarosz
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-348, Krakow, Poland
| | - Anna Woźniak
- Central Forensic Laboratory of the Police, Aleje Ujazdowskie 7, 00-583, Warsaw, Poland
| | - Marta Wojtas
- Jagiellonian University Medical College, Faculty of Medicine, Department of Forensic Medicine, Grzegórzecka 16, 31-531, Krakow, Poland
| | - Christopher Phillips
- Forensic Genetics Unit, Institute of Forensic Sciences, University of Santiago de Compostela, R/ San Francisco s/n, 15782, Santiago de Compostela, Spain
| | - Manfred Kayser
- Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Walther Parson
- Institute of Legal Medicine, Medical University of Innsbruck, Muellerstrasse 44, 6020, Innsbruck, Austria
- Forensic Science Program, The Pennsylvania State University, 13 Thomas Building, University Park, PA, 16802, USA
| | - Wojciech Branicki
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-348, Krakow, Poland.
- Central Forensic Laboratory of the Police, Aleje Ujazdowskie 7, 00-583, Warsaw, Poland.
| |
Collapse
|
33
|
Lancaster EE, Lapato DM, Jackson-Cook C, Strauss JF, Roberson-Nay R, York TP. Maternal biological age assessed in early pregnancy is associated with gestational age at birth. Sci Rep 2021; 11:15440. [PMID: 34326348 PMCID: PMC8322056 DOI: 10.1038/s41598-021-94281-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
Maternal age is an established predictor of preterm birth independent of other recognized risk factors. The use of chronological age makes the assumption that individuals age at a similar rate. Therefore, it does not capture interindividual differences that may exist due to genetic background and environmental exposures. As a result, there is a need to identify biomarkers that more closely index the rate of cellular aging. One potential candidate is biological age (BA) estimated by the DNA methylome. This study investigated whether maternal BA, estimated in either early and/or late pregnancy, predicts gestational age at birth. BA was estimated from a genome-wide DNA methylation platform using the Horvath algorithm. Linear regression methods assessed the relationship between BA and pregnancy outcomes, including gestational age at birth and prenatal perceived stress, in a primary and replication cohort. Prenatal BA estimates from early pregnancy explained variance in gestational age at birth above and beyond the influence of other recognized preterm birth risk factors. Sensitivity analyses indicated that this signal was driven primarily by self-identified African American participants. This predictive relationship was sensitive to small variations in the BA estimation algorithm. Benefits and limitations of using BA in translational research and clinical applications for preterm birth are considered.
Collapse
Affiliation(s)
- Eva E Lancaster
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, 23220, USA.
| | - Dana M Lapato
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Colleen Jackson-Cook
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, 23220, USA.,Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Jerome F Strauss
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, 23220, USA.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Roxann Roberson-Nay
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, 23220, USA.,Department of Psychology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Timothy P York
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, 23220, USA.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| |
Collapse
|
34
|
Wilson SL, Wallingford M. Epigenetic regulation of reproduction in human and in animal models. Mol Hum Reprod 2021; 27:6329199. [PMID: 34318322 DOI: 10.1093/molehr/gaab041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/07/2021] [Indexed: 12/24/2022] Open
Affiliation(s)
- Samantha L Wilson
- Princess Margaret Cancer Centre, University Health Network, Toronto Medical Discovery Tower, Toronto, ON, Canada
| | - Mary Wallingford
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, USA.,Division of Obstetrics and Gynecology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
35
|
Noroozi R, Ghafouri-Fard S, Pisarek A, Rudnicka J, Spólnicka M, Branicki W, Taheri M, Pośpiech E. DNA methylation-based age clocks: From age prediction to age reversion. Ageing Res Rev 2021; 68:101314. [PMID: 33684551 DOI: 10.1016/j.arr.2021.101314] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/25/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
Aging as an irretrievable occurrence throughout the entire life is characterized by a progressive decline in physiological functionality and enhanced disease vulnerability. Numerous studies have demonstrated that epigenetic modifications, particularly DNA methylation (DNAm), correlate with aging and age-related diseases. Several investigations have attempted to predict chronological age using the age-related alterations in the DNAm of certain CpG sites. Here we categorize different studies that tracked the aging process in the DNAm landscape to show how epigenetic age clocks evolved from a chronological age estimator to an indicator of lifespan and healthspan. We also describe the health and disease predictive potential of estimated epigenetic age acceleration regarding different clinical conditions and lifestyle factors. Considering the revealed age-related epigenetic changes, the recent age-reprogramming strategies are discussed which are promising methods for resetting the aging clocks.
Collapse
Affiliation(s)
- Rezvan Noroozi
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aleksandra Pisarek
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Joanna Rudnicka
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | | | - Wojciech Branicki
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ewelina Pośpiech
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
36
|
Matías-García PR, Ward-Caviness CK, Raffield LM, Gao X, Zhang Y, Wilson R, Gào X, Nano J, Bostom A, Colicino E, Correa A, Coull B, Eaton C, Hou L, Just AC, Kunze S, Lange L, Lange E, Lin X, Liu S, Nwanaji-Enwerem JC, Reiner A, Shen J, Schöttker B, Vokonas P, Zheng Y, Young B, Schwartz J, Horvath S, Lu A, Whitsel EA, Koenig W, Adamski J, Winkelmann J, Brenner H, Baccarelli AA, Gieger C, Peters A, Franceschini N, Waldenberger M. DNAm-based signatures of accelerated aging and mortality in blood are associated with low renal function. Clin Epigenetics 2021; 13:121. [PMID: 34078457 PMCID: PMC8170969 DOI: 10.1186/s13148-021-01082-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 04/18/2021] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The difference between an individual's chronological and DNA methylation predicted age (DNAmAge), termed DNAmAge acceleration (DNAmAA), can capture life-long environmental exposures and age-related physiological changes reflected in methylation status. Several studies have linked DNAmAA to morbidity and mortality, yet its relationship with kidney function has not been assessed. We evaluated the associations between seven DNAm aging and lifespan predictors (as well as GrimAge components) and five kidney traits (estimated glomerular filtration rate [eGFR], urine albumin-to-creatinine ratio [uACR], serum urate, microalbuminuria and chronic kidney disease [CKD]) in up to 9688 European, African American and Hispanic/Latino individuals from seven population-based studies. RESULTS We identified 23 significant associations in our large trans-ethnic meta-analysis (p < 1.43E-03 and consistent direction of effect across studies). Age acceleration measured by the Extrinsic and PhenoAge estimators, as well as Zhang's 10-CpG epigenetic mortality risk score (MRS), were associated with all parameters of poor kidney health (lower eGFR, prevalent CKD, higher uACR, microalbuminuria and higher serum urate). Six of these associations were independently observed in European and African American populations. MRS in particular was consistently associated with eGFR (β = - 0.12, 95% CI = [- 0.16, - 0.08] change in log-transformed eGFR per unit increase in MRS, p = 4.39E-08), prevalent CKD (odds ratio (OR) = 1.78 [1.47, 2.16], p = 2.71E-09) and higher serum urate levels (β = 0.12 [0.07, 0.16], p = 2.08E-06). The "first-generation" clocks (Hannum, Horvath) and GrimAge showed different patterns of association with the kidney traits. Three of the DNAm-estimated components of GrimAge, namely adrenomedullin, plasminogen-activation inhibition 1 and pack years, were positively associated with higher uACR, serum urate and microalbuminuria. CONCLUSION DNAmAge acceleration and DNAm mortality predictors estimated in whole blood were associated with multiple kidney traits, including eGFR and CKD, in this multi-ethnic study. Epigenetic biomarkers which reflect the systemic effects of age-related mechanisms such as immunosenescence, inflammaging and oxidative stress may have important mechanistic or prognostic roles in kidney disease. Our study highlights new findings linking kidney disease to biological aging, and opportunities warranting future investigation into DNA methylation biomarkers for prognostic or risk stratification in kidney disease.
Collapse
Affiliation(s)
- Pamela R Matías-García
- TUM School of Medicine, Technical University of Munich, Munich, Germany.
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany.
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| | - Cavin K Ward-Caviness
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, NC, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Xu Gao
- Laboratory of Precision Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Yan Zhang
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rory Wilson
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany
| | - Xīn Gào
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jana Nano
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Andrew Bostom
- Center For Primary Care and Prevention, Memorial Hospital of Rhode Island, Pawtucket, RI, USA
| | - Elena Colicino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adolfo Correa
- Departments of Medicine and Pediatrics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Brent Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Charles Eaton
- Center For Primary Care and Prevention, Memorial Hospital of Rhode Island, Pawtucket, RI, USA
- Department of Family Medicine, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Lifang Hou
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Allan C Just
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sonja Kunze
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany
| | - Leslie Lange
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Ethan Lange
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Xihong Lin
- Veterans Affairs Normative Aging Study, Veterans Affairs Boston Healthcare System, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Simin Liu
- Department of Epidemiology, School of Public Health, Brown University, Providence, RI, USA
| | | | - Alex Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Jincheng Shen
- Department of Population Health Sciences, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Pantel Vokonas
- Veterans Affairs Normative Aging Study, Veterans Affairs Boston Healthcare System, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Yinan Zheng
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Bessie Young
- Nephrology, Hospital and Specialty Medicine and Center for Innovation for Veteran-Centered and Value Driven Care, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, WA, USA
| | - Joel Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ake Lu
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Wolfgang Koenig
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Jerzy Adamski
- Research Unit Molecular Endocrinology and Metabolism, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany
- Chair for Experimental Genetics, Technical University of Munich, Freising-Weihenstephan, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany
- Chair Neurogenetics, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Andrea A Baccarelli
- Laboratory of Precision Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Christian Gieger
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany.
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich/Neuherberg, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
37
|
Rackova L, Mach M, Brnoliakova Z. An update in toxicology of ageing. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 84:103611. [PMID: 33581363 DOI: 10.1016/j.etap.2021.103611] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/17/2021] [Accepted: 02/03/2021] [Indexed: 06/12/2023]
Abstract
The field of ageing research has been rapidly advancing in recent decades and it had provided insight into the complexity of ageing phenomenon. However, as the organism-environment interaction appears to significantly affect the organismal pace of ageing, the systematic approach for gerontogenic risk assessment of environmental factors has yet to be established. This puts demand on development of effective biomarker of ageing, as a relevant tool to quantify effects of gerontogenic exposures, contingent on multidisciplinary research approach. Here we review the current knowledge regarding the main endogenous gerontogenic pathways involved in acceleration of ageing through environmental exposures. These include inflammatory and oxidative stress-triggered processes, dysregulation of maintenance of cellular anabolism and catabolism and loss of protein homeostasis. The most effective biomarkers showing specificity and relevancy to ageing phenotypes are summarized, as well. The crucial part of this review was dedicated to the comprehensive overview of environmental gerontogens including various types of radiation, certain types of pesticides, heavy metals, drugs and addictive substances, unhealthy dietary patterns, and sedentary life as well as psychosocial stress. The reported effects in vitro and in vivo of both recognized and potential gerontogens are described with respect to the up-to-date knowledge in geroscience. Finally, hormetic and ageing decelerating effects of environmental factors are briefly discussed, as well.
Collapse
Affiliation(s)
- Lucia Rackova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska cesta 9, 841 04 Bratislava, Slovakia.
| | - Mojmir Mach
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska cesta 9, 841 04 Bratislava, Slovakia
| | - Zuzana Brnoliakova
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska cesta 9, 841 04 Bratislava, Slovakia
| |
Collapse
|
38
|
Wolf EJ, Chen CD, Zhao X, Zhou Z, Morrison FG, Daskalakis NP, Stone A, Schichman S, Grenier JG, Fein-Schaffer D, Huber BR, Abraham CR, Miller MW, Logue MW. Klotho, PTSD, and advanced epigenetic age in cortical tissue. Neuropsychopharmacology 2021; 46:721-730. [PMID: 33096543 PMCID: PMC8027437 DOI: 10.1038/s41386-020-00884-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/12/2020] [Accepted: 09/29/2020] [Indexed: 01/04/2023]
Abstract
This study examined the klotho (KL) longevity gene polymorphism rs9315202 and psychopathology, including posttraumatic stress disorder (PTSD), depression, and alcohol-use disorders, in association with advanced epigenetic age in three postmortem cortical tissue regions: dorsolateral and ventromedial prefrontal cortices and motor cortex. Using data from the VA National PTSD Brain Bank (n = 117), we found that rs9315202 interacted with PTSD to predict advanced epigenetic age in motor cortex among the subset of relatively older (>=45 years), white non-Hispanic decedents (corrected p = 0.014, n = 42). An evaluation of 211 additional common KL variants revealed that only variants in linkage disequilibrium with rs9315202 showed similarly high levels of significance. Alcohol abuse was nominally associated with advanced epigenetic age in motor cortex (p = 0.039, n = 114). The rs9315202 SNP interacted with PTSD to predict decreased KL expression via DNAm age residuals in motor cortex among older white non-Hispanics decedents (indirect β = -0.198, p = 0.027). Finally, in dual-luciferase enhancer reporter system experiments, we found that inserting the minor allele of rs9315202 in a human kidney cell line HK-2 genomic DNA resulted in a change in KL transcriptional activities, likely operating via long noncoding RNA in this region. This was the first study to examine multiple forms of psychopathology in association with advanced DNA methylation age across several brain regions, to extend work concerning the association between rs9315202 and advanced epigenetic to brain tissue, and to identify the effects of rs9315202 on KL gene expression. KL augmentation holds promise as a therapeutic intervention to slow the pace of cellular aging, disease onset, and neuropathology, particularly in older, stressed populations.
Collapse
Affiliation(s)
- Erika J Wolf
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA.
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA.
| | - Ci-Di Chen
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Xiang Zhao
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Zhenwei Zhou
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Filomene G Morrison
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | | | - Annjanette Stone
- Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Steven Schichman
- Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Jaclyn Garza Grenier
- Brigham and Women's Hospital, Channing Division of Network Medicine, Boston, MA, USA
| | - Dana Fein-Schaffer
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA
| | - Bertrand R Huber
- Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Carmela R Abraham
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Mark W Miller
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Mark W Logue
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Biomedical Genetics, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
39
|
Nwanaji-Enwerem JC, Colicino E. DNA Methylation-Based Biomarkers of Environmental Exposures for Human Population Studies. Curr Environ Health Rep 2021; 7:121-128. [PMID: 32062850 DOI: 10.1007/s40572-020-00269-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This manuscript orients the reader to the underlying motivations of environmental biomarker development for human population studies and provides the foundation for applying these novel biomarkers in future research. In this review, we focus our attention on the DNA methylation-based biomarkers of (i) smoking, among adults and pregnant women, (ii) lifetime cannabis use, (iii) alcohol consumption, and (iv) cumulative exposure to lead. RECENT FINDINGS Prior environmental exposures and lifestyle modulate DNA methylation levels. Exposure-related DNA methylation changes can either be persistent or reversible once the exposure is no longer present, and this combination of both persistent and reversible changes has essential value for biomarker development. Here, we present available biomarkers representing past and cumulative exposures using individual DNA methylation profiles. In the present work, we describe how the field of environmental epigenetics can leverage machine learning algorithms to develop exposure biomarkers and reduce problems of misreporting exposures or limited access technology. We emphasize the crucial role of the individual DNA methylation profiles in those predictions, providing a summary of each biomarker, and highlighting their advantages, and limitations. Future research can cautiously leverage these DNA methylation-based biomarkers to understand the onset and progression of diseases.
Collapse
Affiliation(s)
- Jamaji C Nwanaji-Enwerem
- Belfer Center for Science and International Affairs, Harvard Kennedy School of Government, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Elena Colicino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 17 E 102nd St. West 3rd Floor, New York, NY, 10029, USA.
| |
Collapse
|
40
|
Odintsova VV, Rebattu V, Hagenbeek FA, Pool R, Beck JJ, Ehli EA, van Beijsterveldt CEM, Ligthart L, Willemsen G, de Geus EJC, Hottenga JJ, Boomsma DI, van Dongen J. Predicting Complex Traits and Exposures From Polygenic Scores and Blood and Buccal DNA Methylation Profiles. Front Psychiatry 2021; 12:688464. [PMID: 34393852 PMCID: PMC8357987 DOI: 10.3389/fpsyt.2021.688464] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/15/2021] [Indexed: 11/13/2022] Open
Abstract
We examined the performance of methylation scores (MS) and polygenic scores (PGS) for birth weight, BMI, prenatal maternal smoking exposure, and smoking status to assess the extent to which MS could predict these traits and exposures over and above the PGS in a multi-omics prediction model. MS may be seen as the epigenetic equivalent of PGS, but because of their dynamic nature and sensitivity of non-genetic exposures may add to complex trait prediction independently of PGS. MS and PGS were calculated based on genotype data and DNA-methylation data in blood samples from adults (Illumina 450 K; N = 2,431; mean age 35.6) and in buccal samples from children (Illumina EPIC; N = 1,128; mean age 9.6) from the Netherlands Twin Register. Weights to construct the scores were obtained from results of large epigenome-wide association studies (EWASs) based on whole blood or cord blood methylation data and genome-wide association studies (GWASs). In adults, MSs in blood predicted independently from PGSs, and outperformed PGSs for BMI, prenatal maternal smoking, and smoking status, but not for birth weight. The largest amount of variance explained by the multi-omics prediction model was for current vs. never smoking (54.6%) of which 54.4% was captured by the MS. The two predictors captured 16% of former vs. never smoking initiation variance (MS:15.5%, PGS: 0.5%), 17.7% of prenatal maternal smoking variance (MS:16.9%, PGS: 0.8%), 11.9% of BMI variance (MS: 6.4%, PGS 5.5%), and 1.9% of birth weight variance (MS: 0.4%, PGS: 1.5%). In children, MSs in buccal samples did not show independent predictive value. The largest amount of variance explained by the two predictors was for prenatal maternal smoking (2.6%), where the MSs contributed 1.5%. These results demonstrate that blood DNA MS in adults explain substantial variance in current smoking, large variance in former smoking, prenatal smoking, and BMI, but not in birth weight. Buccal cell DNA methylation scores have lower predictive value, which could be due to different tissues in the EWAS discovery studies and target sample, as well as to different ages. This study illustrates the value of combining polygenic scores with information from methylation data for complex traits and exposure prediction.
Collapse
Affiliation(s)
- Veronika V Odintsova
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Valerie Rebattu
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Fiona A Hagenbeek
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - René Pool
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jeffrey J Beck
- Avera Institute for Human Genetics, Sioux Falls, SD, United States
| | - Erik A Ehli
- Avera Institute for Human Genetics, Sioux Falls, SD, United States
| | - Catharina E M van Beijsterveldt
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Lannie Ligthart
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Gonneke Willemsen
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Eco J C de Geus
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jenny van Dongen
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
41
|
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: 2.3] [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
|
42
|
Ryan J, Wrigglesworth J, Loong J, Fransquet PD, Woods RL. A Systematic Review and Meta-analysis of Environmental, Lifestyle, and Health Factors Associated With DNA Methylation Age. J Gerontol A Biol Sci Med Sci 2020; 75:481-494. [PMID: 31001624 DOI: 10.1093/gerona/glz099] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Indexed: 02/07/2023] Open
Abstract
DNA methylation (DNAm) algorithms of biological age provide a robust estimate of an individual's chronological age and can predict their risk of age-related disease and mortality. This study reviewed the evidence that environmental, lifestyle and health factors are associated with the Horvath and Hannum epigenetic clocks. A systematic search identified 61 studies. Chronological age was correlated with DNAm age in blood (median .83, range .13-.99). In a meta-analysis body mass index (BMI) was associated with increased DNAm age (Hannum β: 0.07, 95% CI 0.04 to 0.10; Horvath β: 0.06, 95% CI 0.02 to 0.10), but there was no association with smoking (Hannum β: 0.12, 95% CI -0.50 to 0.73; Horvath β:0.18, 95% CI -0.10 to 0.46). DNAm age was positively associated with frailty (three studies, n = 3,093), and education was negatively associated with the Hannum estimate of DNAm age specifically (four studies, n = 13,955). For most other exposures, findings were too inconsistent to draw conclusions. In conclusion, BMI was positively associated with biological aging measured using DNAm, with some evidence that frailty also increased aging. More research is needed to provide conclusive evidence regarding other exposures. This field of research has the potential to provide further insights into how to promote slower biological aging and ultimately prolong healthy life.
Collapse
Affiliation(s)
- Joanne Ryan
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia.,INSERM, Univ Montpellier, Neuropsychiatry, Epidemiological and Clinical Research, Montpellier, France
| | - Jo Wrigglesworth
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Jun Loong
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Peter D Fransquet
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Robyn L Woods
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
43
|
He X, Liu J, Liu B, Shi J. The use of DNA methylation clock in aging research. Exp Biol Med (Maywood) 2020; 246:436-446. [PMID: 33175612 DOI: 10.1177/1535370220968802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
One of the key characteristics of aging is a progressive loss of physiological integrity, which weakens bodily functions and increases the risk of death. A robust biomarker is important for the assessment of biological age, the rate of aging, and a person's health status. DNA methylation clocks, novel biomarkers of aging, are composed of a group of cytosine-phosphate-guanine dinucleotides, the DNA methylation status of which can be used to accurately measure subjective age. These clocks are considered accurate biomarkers of chronological age for humans and other vertebrates. Numerous studies have demonstrated these clocks to quantify the rate of biological aging and the effects of longevity and anti-aging interventions. In this review, we describe the purpose and use of DNA methylation clocks in aging research.
Collapse
Affiliation(s)
- Xi He
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, 66367Zunyi Medical University, Zunyi 563003, China
| | - Jiaojiao Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, 66367Zunyi Medical University, Zunyi 563003, China
| | - Bo Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, 66367Zunyi Medical University, Zunyi 563003, China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, 66367Zunyi Medical University, Zunyi 563003, China
| |
Collapse
|
44
|
Shu C, Justice AC, Zhang X, Marconi VC, Hancock DB, Johnson EO, Xu K. DNA methylation biomarker selected by an ensemble machine learning approach predicts mortality risk in an HIV-positive veteran population. Epigenetics 2020; 16:741-753. [PMID: 33092459 PMCID: PMC8216205 DOI: 10.1080/15592294.2020.1824097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Background: With the improved life expectancy of people living with HIV (PLWH), identifying vulnerable subpopulations at high mortality risk is important. Evidences showed that DNA methylation (DNAm) is associated with mortality in non-HIV populations. Here, we established a panel of DNAm biomarkers that can predict mortality risk among PLWH. Methods: 1,081 HIV-positive participants from the Veterans Ageing Cohort Study (VACS) were divided into training (N = 460), validation (N = 114), and testing (N = 507) sets. VACS index was used as a measure of mortality risk among PLWH. Model training and fine-tuning were conducted using the ensemble method in the training and validation sets and prediction performance was assessed in the testing set. The survival analysis comparing the predicted high and low mortality risk groups and the Gene Ontology enrichment analysis of the predictive CpG sites were performed. Results: We selected a panel of 393 CpGs for the ensemble prediction model that showed excellent performance in predicting high mortality risk with an auROC of 0.809 (95%CI: 0.767,0.851) and a balanced accuracy of 0.653 (95%CI: 0.611, 0.693) in the testing set. The high mortality risk group was significantly associated with 10-year mortality (hazard ratio = 1.79, p = 4E-05) compared with low risk group. These 393 CpGs were located in 280 genes enriched in immune and inflammation response pathways. Conclusions: We identified a panel of DNAm features associated with mortality risk in PLWH. These DNAm features may serve as predictive biomarkers for mortality risk among PLWH. Abbreviations: AUC: Area Under Curve; CI: Confidence interval; DMR: differentially methylated region; DNA: Deoxyribonucleic acid; DNAm: DNA methylation; DAVID: Database for Annotation, Visualization, and Integrated Discovery; EWA: epigenome-wide association; FDR: False discovery rate; FWER: Family-wise error rate; GLMNET: elastic-net-regularized generalized linear models; GO: Gene ontology; HIV: Human immunodeficiency virus; HM450K: Human Methylation 450 K BeadChip; k-NN: k-nearest neighbours; NK: Natural killer; PC: Principal component; PLWH: people living with HIV; QC: Quality control; SVM: Support Vector Machines; VACS: Veterans Ageing Cohort Study; XGBoost: Extreme Gradient Boosting Tree
Collapse
Affiliation(s)
- Chang Shu
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.,Connecticut Veteran Healthcare System, West Haven, CT, USA
| | - Amy C Justice
- Connecticut Veteran Healthcare System, West Haven, CT, USA.,Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Xinyu Zhang
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.,Connecticut Veteran Healthcare System, West Haven, CT, USA
| | - Vincent C Marconi
- Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Dana B Hancock
- GenOmics, Bioinformatics, and Translational Research Center, Biostatistics and Epidemiology Division, RTI International, Research Triangle Park, NC, USA
| | - Eric O Johnson
- GenOmics, Bioinformatics, and Translational Research Center, Biostatistics and Epidemiology Division, RTI International, Research Triangle Park, NC, USA.,Fellow Program, RTI International, Research Triangle Park, NC, USA
| | - Ke Xu
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.,Connecticut Veteran Healthcare System, West Haven, CT, USA
| |
Collapse
|
45
|
Galloway JE, Holderbaum AM, Arya N, Zhang S, Bodnar MS, Norman R, Carson WE, Yu L, Kendra KL, Burd CE. Impact of age-related T cell dynamics on the identification of biomarkers predictive of immunotherapy discontinuation: a prospective cohort study. ACTA ACUST UNITED AC 2020; 1:58-70. [PMID: 34337428 DOI: 10.1002/aac2.12012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background The impact of biologic aging on immune checkpoint inhibitor (ICI) toxicity and efficacy is underexplored in metastatic melanoma (MM). In peripheral blood T-lymphocytes (PBTLs), biologic aging is characterized by changes in T-cell composition and cellular senescence. Whether indicators of PBTL biologic aging vary in MM patients or can be used to predict premature ICI discontinuation (pID) is unknown. Methods We prospectively collected PBTLs from 117 cancer-free controls and 46 MM patients scheduled to begin pembrolizumab or nivolumab monotherapy. 74 mRNAs indicative of T-cell subsets, activation, co-stimuation/inhibition and cellular senescence were measured by Nanostring. Relationships between each mRNA and chronologic age were assessed in patients and controls. Candidate biomarkers were identified by calculating the hazard ratio (HR) for pID in patients divided into low and high groups based on log-transformed mRNA levels or the magnitude by which each mRNA measurement deviated from the control trend (Δage). Area under the curve (AUC) analyses explored the ability of each biomarker to discriminate between patients with and without pID at 6 months and 1 year. Results Fifteen mRNAs correlated with chronologic age in controls, including markers of T-cell subsets, differentiation, cytokine production and co-stimulation/inhibition. None of these mRNAs remained correlated with age in patients. Median follow-up was 94.8 (1.6-195.7) weeks and 35 of 46 patients discontinued therapy (23 progression, 7 toxicity, 5 comorbidity/patient preference). Elevated pre-therapy CD8A (HR 2.2[1.1-4.9]), CD45RB (HR 2.9[1.4-5.8]) and TNFRSF14 (HR 2.2[1.1-4.5]) levels predicted pID independent of Δage-correction. CD3ε, CD27 and FOXO1 predicted pID only after Δage-correction (HR 2.5[1.3-5.1]; 3.7[1.8-7.8]; 2.1[1.1-4.3]). AUC analysis identified Δage-CD3ε and -CD27 as candidate predictors of pID (AUC=0.73; 0.75). Conclusions Correlations between transcriptional markers of PBTL composition and chronologic age are disrupted in MM. Correcting for normal, age-related trends in biomarker expression unveils new biomarker candidates predictive of ICI outcomes.
Collapse
Affiliation(s)
- Jason E Galloway
- Department of Molecular Genetics, The Ohio State University College of Arts and Sciences, Columbus, OH 43210.,Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Andrea M Holderbaum
- Department of Molecular Genetics, The Ohio State University College of Arts and Sciences, Columbus, OH 43210.,Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Namrata Arya
- Department of Molecular Genetics, The Ohio State University College of Arts and Sciences, Columbus, OH 43210.,Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Suohui Zhang
- Department of Molecular Genetics, The Ohio State University College of Arts and Sciences, Columbus, OH 43210.,Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Michael S Bodnar
- Department of Molecular Genetics, The Ohio State University College of Arts and Sciences, Columbus, OH 43210.,Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Ruthann Norman
- Department of Surgery, Division of Surgical Oncology, The Ohio State University College of Medicine, Columbus, OH 43210
| | - William E Carson
- Department of Surgery, Division of Surgical Oncology, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Lianbo Yu
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Kari L Kendra
- Medical Oncology Division, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Christin E Burd
- Department of Molecular Genetics, The Ohio State University College of Arts and Sciences, Columbus, OH 43210.,Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210
| |
Collapse
|
46
|
Wagner-Skacel J, Dalkner N, Moerkl S, Kreuzer K, Farzi A, Lackner S, Painold A, Reininghaus EZ, Butler MI, Bengesser S. Sleep and Microbiome in Psychiatric Diseases. Nutrients 2020; 12:nu12082198. [PMID: 32718072 PMCID: PMC7468877 DOI: 10.3390/nu12082198] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/15/2020] [Accepted: 07/20/2020] [Indexed: 12/29/2022] Open
Abstract
Objectives: Disturbances in the gut–brain barrier play an essential role in the development of mental disorders. There is considerable evidence showing that the gut microbiome not only affects digestive, metabolic and immune functions of the host but also regulates host sleep and mental states through the microbiota–gut–brain axis. The present review summarizes the role of the gut microbiome in the context of circadian rhythms, nutrition and sleep in psychiatric disorders. Methods: A PubMed search (studies published between April 2015–April 2020) was conducted with the keywords: “sleep, microbiome and psychiatry”; “sleep, microbiome and depression”; “sleep, microbiome and bipolar disorder”, “sleep, microbiome and schizophrenia”, “sleep, microbiome and anorexia nervosa”, “sleep, microbiome and substance use disorder”, “sleep, microbiome and anxiety”; “clock gene expression and microbiome”, “clock gene expression and nutrition”. Only studies investigating the relationship between sleep and microbiome in psychiatric patients were included in the review. Results: Search results yielded two cross-sectional studies analyzing sleep and gut microbiome in 154 individuals with bipolar disorder and one interventional study analyzing the effect of fecal microbiota transplantation in 17 individuals with irritable bowel syndrome on sleep. In patients with bipolar disorder, Faecalibacterium was significantly associated with improved sleep quality scores and a significant correlation between Lactobacillus counts and sleep. Conclusion: Translational research on this important field is limited and further investigation of the bidirectional pathways on sleep and the gut microbiome in mood disorders is warranted.
Collapse
Affiliation(s)
- Jolana Wagner-Skacel
- Department of Medical Psychology, Medical University of Graz (MUG), 8036 Graz, Austria;
| | - Nina Dalkner
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz (MUG), 8036 Graz, Austria; (N.D.); (S.M.); (K.K.); (A.P.); (E.Z.R.)
| | - Sabrina Moerkl
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz (MUG), 8036 Graz, Austria; (N.D.); (S.M.); (K.K.); (A.P.); (E.Z.R.)
| | - Kathrin Kreuzer
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz (MUG), 8036 Graz, Austria; (N.D.); (S.M.); (K.K.); (A.P.); (E.Z.R.)
| | - Aitak Farzi
- Otto Loewi Research Center (for Vascular Biology, Immunology and Inflammation), Division of Pharmacology, Medical University of Graz (MUG), 8036 Graz, Austria;
| | - Sonja Lackner
- Otto Loewi Research Center (for Vascular Biology, Immunology andI), Division of Immunology and Pathophysiology, Medical University of Graz (MUG), 8036 Graz, Austria;
| | - Annamaria Painold
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz (MUG), 8036 Graz, Austria; (N.D.); (S.M.); (K.K.); (A.P.); (E.Z.R.)
| | - Eva Z. Reininghaus
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz (MUG), 8036 Graz, Austria; (N.D.); (S.M.); (K.K.); (A.P.); (E.Z.R.)
| | - Mary I. Butler
- Department of Psychiatry, University College Cork, T12 YN60 Cork, Ireland;
| | - Susanne Bengesser
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz (MUG), 8036 Graz, Austria; (N.D.); (S.M.); (K.K.); (A.P.); (E.Z.R.)
- Correspondence: ; Tel.: +43-316-86224
| |
Collapse
|
47
|
Koop BE, Reckert A, Becker J, Han Y, Wagner W, Ritz-Timme S. Epigenetic clocks may come out of rhythm-implications for the estimation of chronological age in forensic casework. Int J Legal Med 2020; 134:2215-2228. [PMID: 32661599 PMCID: PMC7578121 DOI: 10.1007/s00414-020-02375-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 07/08/2020] [Indexed: 01/01/2023]
Abstract
There is a growing perception that DNA methylation may be influenced by exogenous and endogenous parameters. Knowledge of these factors is of great relevance for the interpretation of DNA-methylation data for the estimation of chronological age in forensic casework. We performed a literature review to identify parameters, which might be of relevance for the prediction of chronological age based on DNA methylation. The quality of age predictions might particularly be influenced by lifetime adversities (chronic stress, trauma/post-traumatic stress disorder (PTSD), violence, low socioeconomic status/education), cancer, obesity and related diseases, infectious diseases (especially HIV and Cytomegalovirus (CMV) infections), sex, ethnicity and exposure to toxins (alcohol, smoking, air pollution, pesticides). Such factors may alter the DNA methylation pattern and may explain the partly high deviations between epigenetic age and chronological age in single cases (despite of low mean absolute deviations) that can also be observed with “epigenetic clocks” comprising a high number of CpG sites. So far, only few publications dealing with forensic age estimation address these confounding factors. Future research should focus on the identification of further relevant confounding factors and the development of models that are “robust” against the influence of such biological factors by systematic investigations under targeted inclusion of diverse and defined cohorts.
Collapse
Affiliation(s)
- Barbara Elisabeth Koop
- Institute of Legal Medicine, University Hospital Düsseldorf, 40225, Düsseldorf, Germany.
| | - Alexandra Reckert
- Institute of Legal Medicine, University Hospital Düsseldorf, 40225, Düsseldorf, Germany
| | - Julia Becker
- Institute of Legal Medicine, University Hospital Düsseldorf, 40225, Düsseldorf, Germany
| | - Yang Han
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen Faculty of Medicine, Aachen, Germany
| | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen Faculty of Medicine, Aachen, Germany
| | - Stefanie Ritz-Timme
- Institute of Legal Medicine, University Hospital Düsseldorf, 40225, Düsseldorf, Germany
| |
Collapse
|
48
|
Sabi SH, Khabour OF, Alzoubi KH, Cobb CO, Eissenberg T. Changes at global and site-specific DNA methylation of MLH1 gene promoter induced by waterpipe smoking in blood lymphocytes and oral epithelial cells. Inhal Toxicol 2020; 32:124-130. [PMID: 32319830 DOI: 10.1080/08958378.2020.1754972] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective: Cigarette tobacco smoking has been shown to cause cancer through different mechanisms that include epigenetic modulation of tumor-suppressor genes. In the present study, the association between global and MLH1 gene promoter methylation and waterpipe tobacco smoking was investigated. Materials and Methods: Blood lymphocytes and oral epithelium were sampled from 150 pure waterpipe smokers and 150 never-smokers from Jordan. Methylation assessment was performed using the methylation-specific PCR technique for MLH1 gene and ELISA for global DNA methylation. Results: Significant increases were shown in global DNA methylation as measured in blood lymphocytes (p < 0.01). In addition, increases in MLH1 gene promoter methylation among waterpipe smokers compared to nonsmokers (p < 0.001) in both oral epithelium and blood lymphocytes was also observed. In addition, strong correlation was found between LWDS-10J dependence score and magnitude of promoter specific methylation of MLH1 (r2 = 0.74-0.78, p < 0.001). Moreover, the percentage of methylated MLH1 promoter was not affected by age or gender (p > 0.05). Discussion and Conclusion: Collectively, the results indicate that waterpipe tobacco use is associated with epigenetic changes that might predispose users to lung and blood cancers. The results highlight the need for actions to discourage waterpipe smoking and can be used in cessation interventions that target this type of smoking.
Collapse
Affiliation(s)
- Salsabeel H Sabi
- Department of Applied Biology, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F Khabour
- Department of Medical laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H Alzoubi
- Department of Clinical Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Caroline O Cobb
- Department of Psychology and Center for the Study of Tobacco Products, Virginia Commonwealth University, Richmond, VA, USA
| | - Thomas Eissenberg
- Department of Psychology and Center for the Study of Tobacco Products, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
49
|
Lei MK, Gibbons FX, Simons RL, Philibert RA, Beach SRH. The Effect of Tobacco Smoking Differs across Indices of DNA Methylation-Based Aging in an African American Sample: DNA Methylation-Based Indices of Smoking Capture These Effects. Genes (Basel) 2020; 11:E311. [PMID: 32183340 PMCID: PMC7140795 DOI: 10.3390/genes11030311] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/09/2020] [Accepted: 03/12/2020] [Indexed: 01/09/2023] Open
Abstract
Smoking is one of the leading preventable causes of morbidity and mortality worldwide, prompting interest in its association with DNA methylation-based measures of biological aging. Considerable progress has been made in developing DNA methylation-based measures that correspond to self-reported smoking status. In addition, assessment of DNA methylation-based aging has been expanded to better capture individual differences in risk for morbidity and mortality. Untested to date, however, is whether smoking is similarly related to older and newer indices of DNA methylation-based aging, and whether DNA methylation-based indices of smoking can be used in lieu of self-reported smoking to examine effects on DNA methylation-based aging measures. In the current investigation we examine mediation of the impact of self-reported cigarette consumption on accelerated, intrinsic DNA methylation-based aging using indices designed to predict chronological aging, phenotypic aging, and mortality risk, as well as a newly developed DNA methylation-based measure of telomere length. Using a sample of 500 African American middle aged smokers and non-smokers, we found that a) self-reported cigarette consumption was associated with accelerated intrinsic DNA methylation-based aging on some but not all DNA methylation-based aging indices, b) for those aging outcomes associated with self-reported cigarette consumption, DNA methylation-based indicators of smoking typically accounted for greater variance than did self-reported cigarette consumption, and c) self-reported cigarette consumption effects on DNA methylation-based aging indices typically were fully mediated by DNA methylation-based indicators of smoking (e.g., PACKYRS from GrimAge; or cg05575921 CpG site). Results suggest that when DNA methylation-based indices of smoking are substituted for self-reported assessments of smoking, they will typically fully reflect the varied impact of cigarette smoking on intrinsic, accelerated DNA methylation-based aging.
Collapse
Affiliation(s)
- Man-Kit Lei
- Department of Sociology, University of Georgia, Athens, GA 30602, USA; (M.-K.L.); (R.L.S.)
| | - Frederick X. Gibbons
- Department of Psychological Sciences, University of Connecticut, Storrs, CT 06269, USA;
| | - Ronald L. Simons
- Department of Sociology, University of Georgia, Athens, GA 30602, USA; (M.-K.L.); (R.L.S.)
| | - Robert A. Philibert
- Department of Psychiatry, University of Iowa, Iowa, IA 52242, USA;
- Behavioral Diagnostics, Coralville, Iowa, IA 52241, USA
| | - Steven R. H. Beach
- Department of Psychology, University of Georgia, Athens, GA 30602, USA
- Center for Family Research, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
50
|
Logue MW, Miller MW, Wolf EJ, Huber BR, Morrison FG, Zhou Z, Zheng Y, Smith AK, Daskalakis NP, Ratanatharathorn A, Uddin M, Nievergelt CM, Ashley-Koch AE, Baker DG, Beckham JC, Garrett ME, Boks MP, Geuze E, Grant GA, Hauser MA, Kessler RC, Kimbrel NA, Maihofer AX, Marx CE, Qin XJ, Risbrough VB, Rutten BPF, Stein MB, Ursano RJ, Vermetten E, Vinkers CH, Ware EB, Stone A, Schichman SA, McGlinchey RE, Milberg WP, Hayes JP, Verfaellie M. An epigenome-wide association study of posttraumatic stress disorder in US veterans implicates several new DNA methylation loci. Clin Epigenetics 2020; 12:46. [PMID: 32171335 PMCID: PMC7071645 DOI: 10.1186/s13148-020-0820-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/29/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Previous studies using candidate gene and genome-wide approaches have identified epigenetic changes in DNA methylation (DNAm) associated with posttraumatic stress disorder (PTSD). METHODS In this study, we performed an EWAS of PTSD in a cohort of Veterans (n = 378 lifetime PTSD cases and 135 controls) from the Translational Research Center for TBI and Stress Disorders (TRACTS) cohort assessed using the Illumina EPIC Methylation BeadChip which assesses DNAm at more than 850,000 sites throughout the genome. Our model included covariates for ancestry, cell heterogeneity, sex, age, and a smoking score based on DNAm at 39 smoking-associated CpGs. We also examined in EPIC-based DNAm data generated from pre-frontal cortex (PFC) tissue from the National PTSD Brain Bank (n = 72). RESULTS The analysis of blood samples yielded one genome-wide significant association with PTSD at cg19534438 in the gene G0S2 (p = 1.19 × 10-7, padj = 0.048). This association was replicated in an independent PGC-PTSD-EWAS consortium meta-analysis of military cohorts (p = 0.0024). We also observed association with the smoking-related locus cg05575921 in AHRR despite inclusion of a methylation-based smoking score covariate (p = 9.16 × 10-6), which replicates a previously observed PGC-PTSD-EWAS association (Smith et al. 2019), and yields evidence consistent with a smoking-independent effect. The top 100 EWAS loci were then examined in the PFC data. One of the blood-based PTSD loci, cg04130728 in CHST11, which was in the top 10 loci in blood, but which was not genome-wide significant, was significantly associated with PTSD in brain tissue (in blood p = 1.19 × 10-5, padj = 0.60, in brain, p = 0.00032 with the same direction of effect). Gene set enrichment analysis of the top 500 EWAS loci yielded several significant overlapping GO terms involved in pathogen response, including "Response to lipopolysaccharide" (p = 6.97 × 10-6, padj = 0.042). CONCLUSIONS The cross replication observed in independent cohorts is evidence that DNA methylation in peripheral tissue can yield consistent and replicable PTSD associations, and our results also suggest that that some PTSD associations observed in peripheral tissue may mirror associations in the brain.
Collapse
Affiliation(s)
- Mark W. Logue
- grid.410370.10000 0004 4657 1992National Center for PTSD, VA Boston Healthcare System, Boston, MA USA ,grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA ,grid.475010.70000 0004 0367 5222,Biomedical Genetics, Boston University School of Medicine, Boston, MA USA ,grid.189504.10000 0004 1936 7558Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Mark W. Miller
- grid.410370.10000 0004 4657 1992National Center for PTSD, VA Boston Healthcare System, Boston, MA USA ,grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Erika J. Wolf
- grid.410370.10000 0004 4657 1992National Center for PTSD, VA Boston Healthcare System, Boston, MA USA ,grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Bertrand Russ Huber
- grid.410370.10000 0004 4657 1992National Center for PTSD, VA Boston Healthcare System, Boston, MA USA ,grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Filomene G. Morrison
- grid.410370.10000 0004 4657 1992National Center for PTSD, VA Boston Healthcare System, Boston, MA USA ,grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Zhenwei Zhou
- grid.189504.10000 0004 1936 7558Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Yuanchao Zheng
- grid.189504.10000 0004 1936 7558Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Alicia K. Smith
- grid.189967.80000 0001 0941 6502Department of Gynecology and Obstetrics, Emory University, Atlanta, GA USA ,grid.189967.80000 0001 0941 6502Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA USA
| | - Nikolaos P. Daskalakis
- grid.38142.3c000000041936754XDepartment of Psychiatry, Harvard Medical School, Boston, MA USA ,grid.240206.20000 0000 8795 072XMcLean Hospital, Belmont, MA USA ,Cohen Veterans Bioscience, Cambridge, MA USA ,grid.59734.3c0000 0001 0670 2351Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Andrew Ratanatharathorn
- grid.21729.3f0000000419368729Department of Epidemiology, Columbia University, New York, NY USA
| | - Monica Uddin
- grid.170693.a0000 0001 2353 285XGenomics Program, University of South Florida College of Public Health, Tampa, FL USA ,grid.170693.a0000 0001 2353 285X,Global Health and Infectious Disease Research Program, University of South Florida College of Public Health, Tampa, FL USA
| | - Caroline M. Nievergelt
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA USA ,grid.410371.00000 0004 0419 2708Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA USA
| | - Allison E. Ashley-Koch
- grid.189509.c0000000100241216Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC USA
| | - Dewleen G. Baker
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA USA ,grid.410371.00000 0004 0419 2708Psychiatry Service, Veterans Affairs San Diego Healthcare System, San Diego, CA USA
| | - Jean C. Beckham
- grid.26009.3d0000 0004 1936 7961Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC USA ,grid.410332.70000 0004 0419 9846Research, Durham VA Medical Center, Durham, NC USA ,grid.281208.10000 0004 0419 3073Genetics Research Laboratory, VA Mid-Atlantic Mental Illness Research, Education, and Clinical Center (MIRECC), Durham, NC USA
| | - Melanie E. Garrett
- grid.189509.c0000000100241216Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC USA
| | - Marco P. Boks
- grid.7692.a0000000090126352Department of Psychiatry, UMC Utrecht Brain Center, Utrecht, Utrecht Netherlands
| | - Elbert Geuze
- grid.7692.a0000000090126352Department of Psychiatry, UMC Utrecht Brain Center, Utrecht, Utrecht Netherlands ,Brain Research and Innovation Centre, Netherlands Ministry of Defence, Utrecht, Utrecht Netherlands
| | - Gerald A. Grant
- grid.240952.80000000087342732Department of Neurosurgery, Stanford University Medical Center, Stanford, CA USA
| | - Michael A. Hauser
- grid.189509.c0000000100241216Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC USA
| | - Ronald C. Kessler
- grid.38142.3c000000041936754XDepartment of Health Care Policy, Harvard Medical School, Boston, MA USA
| | - Nathan A. Kimbrel
- grid.410332.70000 0004 0419 9846Research, Durham VA Medical Center, Durham, NC USA ,grid.281208.10000 0004 0419 3073Genetics Research Laboratory, VA Mid-Atlantic Mental Illness Research, Education, and Clinical Center (MIRECC), Durham, NC USA ,grid.26009.3d0000 0004 1936 7961Duke Molecular Physiology Institute, Duke University, Durham, NC USA
| | - Adam X. Maihofer
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA USA ,grid.410371.00000 0004 0419 2708Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA USA
| | - Christine E. Marx
- grid.21925.3d0000 0004 1936 9000Department of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, Pittsburgh, PA USA ,grid.189509.c0000000100241216Department of Psychiatry & Behavioral Sciences, Duke University Medical Center, Durham, NC USA
| | - Xue-Jun Qin
- grid.189509.c0000000100241216Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC USA
| | - Victoria B. Risbrough
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA USA ,grid.410371.00000 0004 0419 2708Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA USA
| | - Bart P. F. Rutten
- grid.412966.e0000 0004 0480 1382School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht Universitair Medisch Centrum, Maastricht, Limburg Netherlands
| | - Murray B. Stein
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Psychiatry Service, Veterans Affairs San Diego Healthcare System, San Diego, CA USA ,grid.410371.00000 0004 0419 2708Million Veteran Program, Veterans Affairs San Diego Healthcare System, San Diego, CA USA
| | - Robert J. Ursano
- grid.265436.00000 0001 0421 5525Department of Psychiatry, Uniformed Services University, Bethesda, MD USA
| | - Eric Vermetten
- Arq, Psychotrauma Reseach Expert Group, Diemen, NH Netherlands ,grid.10419.3d0000000089452978Department of Psychiatry, Leiden University Medical Center, Leiden, ZH Netherlands ,Netherlands Defense Department, Research Center, Utrecht, UT Netherlands ,grid.137628.90000 0004 1936 8753Department of Psychiatry, New York University School of Medicine, New York, NY USA
| | - Christiaan H. Vinkers
- Department of Anatomy and Neurosciences, Amsterdam UMC (location VUmc), Amsterdam, Holland Netherlands ,Department of Psychiatry, Amsterdam UMC (location VUmc), Amsterdam, Holland Netherlands
| | - Erin B. Ware
- grid.214458.e0000000086837370Institute for Social Research, Survey Research Center, University of Michigan, Michigan, MI USA
| | - Annjanette Stone
- grid.413916.80000 0004 0419 1545Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System, Little Rock, AR USA
| | - Steven A. Schichman
- grid.413916.80000 0004 0419 1545Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System, Little Rock, AR USA
| | - Regina E. McGlinchey
- grid.38142.3c000000041936754XDepartment of Psychiatry, Harvard Medical School, Boston, MA USA ,grid.410370.10000 0004 4657 1992Geriatric Research Educational and Clinical Center and Translational Research Center for TBI and Stress Disorders, VA Boston Health Care System, Boston, MA USA
| | - William P. Milberg
- grid.38142.3c000000041936754XDepartment of Psychiatry, Harvard Medical School, Boston, MA USA ,grid.410370.10000 0004 4657 1992Geriatric Research Educational and Clinical Center and Translational Research Center for TBI and Stress Disorders, VA Boston Health Care System, Boston, MA USA
| | - Jasmeet P. Hayes
- grid.410370.10000 0004 4657 1992National Center for PTSD, VA Boston Healthcare System, Boston, MA USA ,grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA ,grid.261331.40000 0001 2285 7943Department of Psychology and Chronic Brain Injury Program, The Ohio State University, Columbus, OH USA
| | - Mieke Verfaellie
- grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA ,grid.475010.70000 0004 0367 5222Memory Disorders Research Center, VA Boston Healthcare System and Boston University School of Medicine, Boston, MA USA
| | | |
Collapse
|