151
|
Carskadon MA, Chappell KR, Barker DH, Hart AC, Dwyer K, Gredvig-Ardito C, Starr C, McGeary JE. A pilot prospective study of sleep patterns and DNA methylation-characterized epigenetic aging in young adults. BMC Res Notes 2019; 12:583. [PMID: 31526398 PMCID: PMC6747743 DOI: 10.1186/s13104-019-4633-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 09/11/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Molecular markers in DNA methylation at a subset of CpG sites are affected by the environment and contribute to biological (epigenetic) age. We hypothesized that shorter sleep duration and possibly irregular sleep would be associated with accelerated epigenetic aging. We examined epigenetic vs. chronological age in 12 young women selected as shorter or longer sleepers studied prospectively across the first 9 weeks of college using a daily online sleep log. Genomic DNA was isolated from two blood samples spanning the interval, and DNA methylation levels were determined and used to measure epigenetic age. RESULTS Epigenetic vs. chronological age differences averaged 2.07 at Time 1 and 1.21 at Time 2. Sleep duration was computed as average daily total sleep time and sleep regularity was indexed using the Sleep Regularity Index. Participants with longer and more regular sleep showed reduced age difference: mean = - 2.48 [95% CI - 6.11; 1.15]; those with shorter and more irregular sleep showed an increased age difference: 3.03 [0.02; 6.03]; and those with either shorter or more irregular sleep averaged no significant change: - 0.49 [- 3.55; 2.56]. These pilot data suggest that short and irregular sleep, even in a young healthy sample, may be associated with accelerated epigenetic aging.
Collapse
Affiliation(s)
- Mary A Carskadon
- EP Bradley Hospital Sleep Research Laboratory, 300 Duncan Drive, Providence, RI, 02906, USA. .,Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Box G-A1, Providence, RI, 02912, USA.
| | - Kenneth R Chappell
- Providence Veterans Affairs Medical Center, 830 Chalkstone Avenue, Providence, RI, 02098, USA
| | - David H Barker
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Box G-A1, Providence, RI, 02912, USA.,Bradley Hasbro Children's Research Center, CoroWest, 1 Hoppin Street, Suite 204, Providence, RI, 20903, USA
| | - Anne C Hart
- Department of Neuroscience and Robert J. & Nancy D. Carney Institute for Brain Science, Brown University, 185 Meeting Street, Providence, RI, 02912, USA
| | - Kayla Dwyer
- Providence Veterans Affairs Medical Center, 830 Chalkstone Avenue, Providence, RI, 02098, USA
| | | | - Caitlyn Starr
- Providence Veterans Affairs Medical Center, 830 Chalkstone Avenue, Providence, RI, 02098, USA
| | - John E McGeary
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Box G-A1, Providence, RI, 02912, USA.,Providence Veterans Affairs Medical Center, 830 Chalkstone Avenue, Providence, RI, 02098, USA
| |
Collapse
|
152
|
Zhao W, Ammous F, Ratliff S, Liu J, Yu M, Mosley TH, Kardia SLR, Smith JA. Education and Lifestyle Factors Are Associated with DNA Methylation Clocks in Older African Americans. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16173141. [PMID: 31466396 PMCID: PMC6747433 DOI: 10.3390/ijerph16173141] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/06/2019] [Accepted: 08/25/2019] [Indexed: 12/12/2022]
Abstract
DNA methylation (DNAm) clocks are important biomarkers of cellular aging and are associated with a variety of age-related chronic diseases and all-cause mortality. Examining the relationship between education and lifestyle risk factors for age-related diseases and multiple DNAm clocks can increase the understanding of how risk factors contribute to aging at the cellular level. This study explored the association between education or lifestyle risk factors for age-related diseases and the acceleration of four DNAm clocks, including intrinsic (IEAA) and extrinsic epigenetic age acceleration (EEAA), PhenoAge acceleration (PhenoAA), and GrimAge acceleration (GrimAA) in the African American participants of the Genetic Epidemiology Network of Arteriopathy. We performed both cross-sectional and longitudinal analyses. In cross-sectional analyses, gender, education, BMI, smoking, and alcohol consumption were all independently associated with GrimAA, whereas only some of them were associated with other clocks. The effect of smoking and education on GrimAA varied by gender. Longitudinal analyses suggest that age and BMI continued to increase GrimAA, and that age and current smoking continued to increase PhenoAA after controlling DNAm clocks at baseline. In conclusion, education and common lifestyle risk factors were associated with multiple DNAm clocks. However, the association with each risk factor varied by clock, which suggests that different clocks may capture adverse effects from different environmental stimuli.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Farah Ammous
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Scott Ratliff
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jiaxuan Liu
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Miao Yu
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas H Mosley
- Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS 39126, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| |
Collapse
|
153
|
Nwanaji-Enwerem JC, Cardenas A, Chai PR, Weisskopf MG, Baccarelli AA, Boyer EW. Relationships of Long-Term Smoking and Moist Snuff Consumption With a DNA Methylation Age Relevant Smoking Index: An Analysis in Buccal Cells. Nicotine Tob Res 2019; 21:1267-1273. [PMID: 30053132 PMCID: PMC6941707 DOI: 10.1093/ntr/nty156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 07/19/2018] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Currently, there is no widely accepted, non-self-report measure that simultaneously reflects smoking behaviors and is molecularly informative of general disease processes. Recently, researchers developed a smoking index (SI) using nucleated blood cells and a multi-tissue DNA methylation-based predictor of chronological age and disease (DNA methylation age [DNAm-age]). To better understand the utility of this novel SI in readily accessible cell types, we used buccal cell DNA methylation to examine SI relationships with long-term tobacco smoking and moist snuff consumption. METHODS We used a publicly available dataset composed of buccal cell DNA methylation values from 120 middle-aged men (40 long-term smokers, 40 moist snuff consumers, and 40 nonsmokers). DNAm-age (353-CpGs) and SI (66-CpGs) were calculated using CpG sites measured using the Illumina HumanMethylation450 BeadChip. We estimated associations of tobacco consumption habits with both SI and DNAm-age using linear regression models adjusted for chronological age, race, and methylation technical covariates. RESULTS In fully adjusted models with nonsmokers as the reference, smoking (β = 1.08, 95% CI = 0.82 to 1.33, p < .0001) but not snuff consumption (β = .06, 95% CI = -0.19 to 0.32, p = .63) was significantly associated with SI. SI was an excellent predictor of smoking versus nonsmoking (area under the curve = 0.92, 95% CI = 0.85 to 0.98). Four DNAm-age CpGs were differentially methylated between smokers and nonsmokers including cg14992253 [EIF3I], which has been previously shown to be differentially methylated with exposure to long-term fine-particle air pollution (PM2.5). CONCLUSIONS The 66-CpG SI appears to be a useful tool for measuring smoking-specific behaviors in buccal cells. Still, further research is needed to broadly confirm our findings and SI relationships with DNAm-age. IMPLICATIONS Our findings demonstrate that this 66-CpG blood-derived SI can reflect long-term tobacco smoking, but not long-term snuff consumption, in buccal cells. This evidence will be useful as the field works to identify an accurate non-self-report smoking biomarker that can be measured in an easily accessible tissue. Future research efforts should focus on (1) optimizing the relationship of the SI with DNAm-age so that the metric can maximize its utility as a tool for understanding general disease processes, and (2) determining normal values for the SI CpGs so that the measure is not as study sample specific.
Collapse
Affiliation(s)
- Jamaji C Nwanaji-Enwerem
- Department of Environmental Health, Harvard T.H. Chan School of Public Health and MD-PhD Program, Harvard Medical School, Boston, MA
| | - Andres Cardenas
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim HealthCare Institute, Boston, MA
| | - Peter R Chai
- Division of Medical Toxicology, Department of Emergency Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Marc G Weisskopf
- Department of Environmental Health and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Columbia Mailman School of Public Health, New York, NY
| | - Edward W Boyer
- Division of Medical Toxicology, Department of Emergency Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
154
|
Curtis SW, Cobb DO, Kilaru V, Terrell ML, Marder ME, Barr DB, Marsit CJ, Marcus M, Conneely KN, Smith AK. Environmental exposure to polybrominated biphenyl (PBB) associates with an increased rate of biological aging. Aging (Albany NY) 2019; 11:5498-5517. [PMID: 31375641 PMCID: PMC6710070 DOI: 10.18632/aging.102134] [Citation(s) in RCA: 14] [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: 05/21/2019] [Accepted: 07/26/2019] [Indexed: 12/13/2022]
Abstract
Advanced age increases risk for cancer, cardiovascular disease, and all-cause mortality. However, people do not age at the same rate, and biological age (frequently measured through DNA methylation) can be older than chronological age. Environmental factors have been associated with the rate of biological aging, but it is not known whether persistent endocrine-disrupting compounds (EDCs) like polybrominated biphenyl (PBB) would associate with age acceleration. Three different epigenetic age acceleration measures (intrinsic, extrinsic, and phenotypic) were calculated from existing epigenetic data in whole blood from a population highly exposed to PBB (N=658). Association between serum PBB concentration and these measures was tested, controlling for sex, lipid levels, and estimated cell type proportions. Higher PBB levels associated with increased age acceleration (intrinsic: β=0.24, 95%CI=0.01-0.46, p = 0.03; extrinsic: β=0.39, 95%CI=0.12-0.65, p = 0.004; and phenotypic: β=0.30, 95%CI=0.05-0.54, p = 0.01). Neither age when exposed to PBB nor sex statistically interacted with PBB to predict age acceleration, but, in stratified analyses, the association between PBB and age acceleration was only in people exposed before finishing puberty and in men. This suggests that EDCs can associate with the biological aging process, and further studies are warranted to investigate other environmental pollutants' effect on aging.
Collapse
Affiliation(s)
- Sarah W. Curtis
- Genetics and Molecular Biology Program, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | - Dawayland O. Cobb
- Department of Gynecology and Obstetrics, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Varun Kilaru
- Department of Gynecology and Obstetrics, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Metrecia L. Terrell
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - M. Elizabeth Marder
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Dana Boyd Barr
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Carmen J. Marsit
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Michele Marcus
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Karen N. Conneely
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Alicia K. Smith
- Department of Gynecology and Obstetrics, School of Medicine, Emory University, Atlanta, GA 30322, USA
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
155
|
Zheng C, Li L, Xu R. Association of Epigenetic Clock with Consensus Molecular Subtypes and Overall Survival of Colorectal Cancer. Cancer Epidemiol Biomarkers Prev 2019; 28:1720-1724. [PMID: 31375479 DOI: 10.1158/1055-9965.epi-19-0208] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/12/2019] [Accepted: 07/26/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Epigenetic clock, or DNA methylation age, has been shown to highly correlate with chronologic age. Epigenetic age acceleration, the difference between DNA methylation age and individual's chronologic age, was observed in colorectal cancer. However, the association of epigenetic age acceleration with colorectal cancer tumor molecular characteristics, clinical characteristics, and patient outcomes has not been systematically investigated. METHODS DNA methylation ages of 345 patients with colorectal cancer from The Cancer Genome Atlas (TCGA) were computed using the Horvath age prediction model. Multivariate linear regression was used to assess the association of epigenetic age acceleration with molecular and clinical features of colorectal cancer, including consensus molecular subtypes (CMS1-CMS4) and tumor stage Cox proportional hazards regression was used to assess the association of epigenetic age acceleration with survival. RESULTS Epigenetic age acceleration is significantly associated with CMS. Compared with CMS2, epigenetic age acceleration for CMS1, CMS3, and CMS4 was 23.90 years [P = 5.55E-11; 95% confidence interval (CI): 17.10-30.69], 9.16 years (P = 5.84E-03; 95% CI: 2.68-15.65), and 6.05 years (P = 2.69E-02; 95% CI: 0.70-11.41), respectively. Furthermore, epigenetic age acceleration is statistically significantly and positively associated with total mortality (HR = 1.97; 95% CI: 1.14-3.39; P = 0.014). CONCLUSIONS Epigenetic age acceleration is associated with colorectal cancer tumor molecular characteristics, and a significant predictor of overall survival of colorectal cancer, along with age and tumor stage. IMPACT Combining information of colonic tissue epigenetic age acceleration and tumor molecular characteristics may improve prognosis prediction in colorectal cancer.
Collapse
Affiliation(s)
- Chunlei Zheng
- Department of Population and Quantitative Health Sciences, Institute of Computational Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Li Li
- Department of Family Medicine, School of Medicine, University of Virginia, Charlottesville, Virginia.
| | - Rong Xu
- Department of Population and Quantitative Health Sciences, Institute of Computational Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
156
|
Abstract
Identifying and validating molecular targets of interventions that extend the human health span and lifespan has been difficult, as most clinical biomarkers are not sufficiently representative of the fundamental mechanisms of ageing to serve as their indicators. In a recent breakthrough, biomarkers of ageing based on DNA methylation data have enabled accurate age estimates for any tissue across the entire life course. These 'epigenetic clocks' link developmental and maintenance processes to biological ageing, giving rise to a unified theory of life course. Epigenetic biomarkers may help to address long-standing questions in many fields, including the central question: why do we age?
Collapse
|
157
|
DNA Methylation Age-Environmental Influences, Health Impacts, and Its Role in Environmental Epidemiology. Curr Environ Health Rep 2019; 5:317-327. [PMID: 30047075 DOI: 10.1007/s40572-018-0203-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW DNA methylation-based aging biomarkers are valuable tools for evaluating the aging process from a molecular perspective. These epigenetic aging biomarkers can be evaluated across the lifespan and are tissue specific. This review examines the literature relating environmental exposures to DNA methylation-based aging biomarkers and also the literature evaluating these biomarkers as predictors of health outcomes. RECENT FINDINGS Multiple studies evaluated the association between air pollution and DNA methylation age and consistently observed that higher exposures are associated with elevated DNA methylation age. Psychosocial exposures, e.g., traumas and adolescent adversity, and infections are also associated with epigenetic aging. DNA methylation age has been repeatedly associated with mortality, cancer, and cognitive impairment. DNA methylation age is responsive to the environment and predictive of health outcomes. Studies are still needed to evaluate whether DNA methylation age acts as a mediator or modifier of environmental health effects and to understand the impact of factors such as race, gender, and genetics.
Collapse
|
158
|
Chon HS, Sehovic M, Marchion D, Walko C, Xiong Y, Extermann M. Biologic Mechanisms Linked to Prognosis in Ovarian Cancer that May Be Affected by Aging. J Cancer 2019; 10:2604-2618. [PMID: 31258768 PMCID: PMC6584919 DOI: 10.7150/jca.29611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 04/27/2019] [Indexed: 12/20/2022] Open
Abstract
The increase of both life expectancy of the Western industrialized population and cancer incidence with aging is expected to result in a rapid expansion of the elderly cancer population, including patients with epithelial ovarian cancer (EOC). Although the survival of patients with EOC has generally improved over the past three decades, this progress has yet to provide benefits for elderly patients. Compared with young age, advanced age has been reported as an adverse prognostic factor influencing EOC. However, contradicting results have been obtained, and the mechanisms underlying this observation are poorly defined. Few papers have been published on the underlying biological mechanisms that might explain this prognosis trend. We provide an extensive review of mechanisms that have been linked to EOC prognosis and/or aging in the published literature and might underlie this relationship in humans.
Collapse
Affiliation(s)
- Hye Sook Chon
- Department of Gynecology Oncology, Moffitt Cancer Center and Research Institute, Tampa FL, USA
- University of South Florida, Tampa FL, USA
| | - Marina Sehovic
- Senior Adult Oncology Program, Moffitt Cancer Center and Research Institute, Tampa FL, USA
- Department of Individualized Cancer Management, Moffitt Cancer Center and Research Institute, Tampa FL, USA
| | - Douglas Marchion
- Department of Pathology, Moffitt Cancer Center and Research Institute, Tampa FL, USA
| | - Christine Walko
- Department of Individualized Cancer Management, Moffitt Cancer Center and Research Institute, Tampa FL, USA
| | - Yin Xiong
- Department of Pathology, Moffitt Cancer Center and Research Institute, Tampa FL, USA
| | - Martine Extermann
- Senior Adult Oncology Program, Moffitt Cancer Center and Research Institute, Tampa FL, USA
- Department of Individualized Cancer Management, Moffitt Cancer Center and Research Institute, Tampa FL, USA
- University of South Florida, Tampa FL, USA
| |
Collapse
|
159
|
Liu Z, Chen BH, Assimes TL, Ferrucci L, Horvath S, Levine ME. The role of epigenetic aging in education and racial/ethnic mortality disparities among older U.S. Women. Psychoneuroendocrinology 2019; 104:18-24. [PMID: 30784901 PMCID: PMC6555423 DOI: 10.1016/j.psyneuen.2019.01.028] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 12/06/2018] [Accepted: 01/30/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Higher mortality experienced by socially disadvantaged groups and/or racial/ethnic minorities is hypothesized to be, at least in part, due to an acceleration of the aging process. Using a new epigenetic aging measure, Levine DNAmAge, this study aimed to investigate whether epigenetic aging accounts for mortality disparities by race/ethnicity and education in a sample of U.S. postmenopausal women. METHODS 1834 participants from an ancillary study (BA23) in the Women's Health Initiative, a national study that recruited postmenopausal women (50-79 years) were included. Over the 22 years of follow-up, 551 women died, and 31,946 person-years were observed. Levine DNAmAge (unit in years) was calculated based on an equation that we previously developed in an independent sample, which incorporates methylation levels at 513 CpG sites. RESULTS As previously reported, non-Hispanic blacks and Hispanics were epigenetically older than non-Hispanic whites of the same chronological age. Similarly, those with less education had older epigenetic ages than expected in the full sample, as well as among non-Hispanic whites and Hispanics, but not among non-Hispanic blacks. Non-Hispanic blacks and those with low education exhibited the greatest risk of mortality. However, this association was partially attenuated when accounting for differences in DNAmAge. Furthermore, formal mediation analysis suggested that DNAmAge partially mediated the mortality increase among non-Hispanic blacks, compared to non-Hispanic whites (proportion mediated, 15.8%, P = 0.002), as well as the mortality increase for those with less than high school education, compared to college educated (proportion mediated, 11.6%, P < 2E-16). CONCLUSIONS Among a group of postmenopausal women, non-Hispanic blacks and those with less education exhibit higher epigenetic aging, which partially accounts for their shorter life expectancies.
Collapse
Affiliation(s)
- Zuyun Liu
- Department of Pathology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Brian H. Chen
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 9009-57088, USA,Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA 90095-7088, USA
| | - Morgan E. Levine
- Department of Pathology, Yale School of Medicine, New Haven, CT 06511, USA,Department of Epidemiology, Yale School of Public Health, New Haven, CT 06511, USA
| |
Collapse
|
160
|
Freudenheim JL, Shields PG, Song MA, Smiraglia D. DNA Methylation and Smoking: Implications for Understanding Effects of Electronic Cigarettes. CURR EPIDEMIOL REP 2019. [DOI: 10.1007/s40471-019-00191-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
161
|
Murabito JM, Zhao Q, Larson MG, Rong J, Lin H, Benjamin EJ, Levy D, Lunetta KL. Measures of Biologic Age in a Community Sample Predict Mortality and Age-Related Disease: The Framingham Offspring Study. J Gerontol A Biol Sci Med Sci 2019; 73:757-762. [PMID: 28977464 DOI: 10.1093/gerona/glx144] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/19/2017] [Indexed: 01/19/2023] Open
Abstract
Background We tested the association of biologic age (BA) measures constructed from different types of biomarkers with mortality and disease in a community-based sample. Methods In Framingham Offspring participants at Exams 7 (1998-2001, mean age 62 ± 10) and 8 (2005-2008, mean age 67 ± 9), we used the Klemera-Doubal method to estimate clinical BA and inflammatory BA and computed the difference (∆age) between BA and CA. Clinical ∆age was computed at Exam 2 (1979-1983, mean age 45 ± 10). At Exam 8, we computed measures of intrinsic and extrinsic epigenetic age. Participants were followed through 2014 for outcomes. Cox proportional hazards models tested the association of each BA estimate with each outcome adjusting for covariates. Results Sample sizes ranged from 2532 to 3417 participants. In multivariable models, each 1-year increase in clinical ∆age at Exam 2 (hazard ratio [HR] = 1.04-1.06, p < 2 × 10-16) and clinical ∆age and inflammatory ∆age at Exam 7 significantly increased the hazards of mortality and incident cardiovascular disease (HR = 1.01-1.05, p < 2 × 10-7), whereas inflammatory ∆age increased the hazards of cancer (HR = 1.01, p < .05). At Exam 8, increased clinical ∆age, inflammatory ∆age, and extrinsic epigenetic age all significantly increased the hazard of mortality (HR = 1.03-1.05, all p < .05); clinical ∆age and inflammatory ∆age increased cardiovascular disease risk (HR = 1.04-1.05, all p < .01); and clinical ∆age increased cancer risk (HR = 1.03, p < .01) when all three BA measures were included in the model. Intrinsic epigenetic age was not significantly associated with any outcome. Conclusions Our findings suggest BA measures may be complementary in predicting risk for mortality and age-related disease.
Collapse
Affiliation(s)
- Joanne M Murabito
- Framingham Heart Study, Massachusetts
- Section of General Internal Medicine, Department of Medicine, Boston University School of Medicine, Massachusetts
| | - Qiang Zhao
- Department of Biostatistics, Boston University School of Public Health, Massachusetts
| | - Martin G Larson
- Framingham Heart Study, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Massachusetts
| | - Jian Rong
- Department of Biostatistics, Boston University School of Public Health, Massachusetts
| | - Honghuang Lin
- Framingham Heart Study, Massachusetts
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Massachusetts
| | - Emelia J Benjamin
- Framingham Heart Study, Massachusetts
- Department of Medicine, Section of Cardiovascular Medicine and Preventive Medicine, Boston University School of Medicine, Massachusetts
- Department of Epidemiology, Boston University School of Public Health, Massachusetts
| | - Daniel Levy
- Framingham Heart Study, Massachusetts
- The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Kathryn L Lunetta
- Framingham Heart Study, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Massachusetts
| |
Collapse
|
162
|
Fransquet PD, Wrigglesworth J, Woods RL, Ernst ME, Ryan J. The epigenetic clock as a predictor of disease and mortality risk: a systematic review and meta-analysis. Clin Epigenetics 2019; 11:62. [PMID: 30975202 PMCID: PMC6458841 DOI: 10.1186/s13148-019-0656-7] [Citation(s) in RCA: 201] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 03/25/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Ageing is one of the principal risk factors for many chronic diseases. However, there is considerable between-person variation in the rate of ageing and individual differences in their susceptibility to disease and death. Epigenetic mechanisms may play a role in human ageing, and DNA methylation age biomarkers may be good predictors of age-related diseases and mortality risk. The aims of this systematic review were to identify and synthesise the evidence for an association between peripherally measured DNA methylation age and longevity, age-related disease, and mortality risk. METHODS A systematic search was conducted in line with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Using relevant search terms, MEDLINE, Embase, Cochrane Central Register of Controlled Trials, and PsychINFO databases were searched to identify articles meeting the inclusion criteria. Studies were assessed for bias using Joanna Briggs Institute critical appraisal checklists. Data was extracted from studies measuring age acceleration as a predictor of age-related diseases, mortality or longevity, and the findings for similar outcomes compared. Using Review Manager 5.3 software, two meta-analyses (one per epigenetic clock) were conducted on studies measuring all-cause mortality. RESULTS Twenty-three relevant articles were identified, including a total of 41,607 participants. Four studies focused on ageing and longevity, 11 on age-related disease (cancer, cardiovascular disease, and dementia), and 11 on mortality. There was some, although inconsistent, evidence for an association between increased DNA methylation age and risk of disease. Meta-analyses indicated that each 5-year increase in DNA methylation age was associated an 8 to 15% increased risk of mortality. CONCLUSION Due to the small number of studies and heterogeneity in study design and outcomes, the association between DNA methylation age and age-related disease and longevity is inconclusive. Increased epigenetic age was associated with mortality risk, but positive publication bias needs to be considered. Further research is needed to determine the extent to which DNA methylation age can be used as a clinical biomarker.
Collapse
Affiliation(s)
- Peter D Fransquet
- Department of Epidemiology and Preventive Medicine, Monash University, ASPREE, Level 5, The Alfred Centre, 99 Commercial Road, Melbourne, Victoria, 3004, Australia.,Disease Epigenetics, Murdoch Childrens Research Institute, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Jo Wrigglesworth
- Department of Epidemiology and Preventive Medicine, Monash University, ASPREE, Level 5, The Alfred Centre, 99 Commercial Road, Melbourne, Victoria, 3004, Australia
| | - Robyn L Woods
- Department of Epidemiology and Preventive Medicine, Monash University, ASPREE, Level 5, The Alfred Centre, 99 Commercial Road, Melbourne, Victoria, 3004, Australia
| | - Michael E Ernst
- Department of Pharmacy Practice and Science, College of Pharmacy, The University of Iowa, Iowa City, IA, USA.,Department of Family Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Joanne Ryan
- Department of Epidemiology and Preventive Medicine, Monash University, ASPREE, Level 5, The Alfred Centre, 99 Commercial Road, Melbourne, Victoria, 3004, Australia. .,Disease Epigenetics, Murdoch Childrens Research Institute, The University of Melbourne, Parkville, Victoria, 3052, Australia. .,INSERM, U1061, Neuropsychiatrie, Recherche Clinique et Epidémiologique, Neuropsychiatry: Research Epidemiological and Clinic, Université Montpellier, 34000, Montpellier, France.
| |
Collapse
|
163
|
DNA Methylation Clocks in Aging: Categories, Causes, and Consequences. Mol Cell 2019; 71:882-895. [PMID: 30241605 DOI: 10.1016/j.molcel.2018.08.008] [Citation(s) in RCA: 369] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/03/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023]
Abstract
Age-associated changes to the mammalian DNA methylome are well documented and thought to promote diseases of aging, such as cancer. Recent studies have identified collections of individual methylation sites whose aggregate methylation status measures chronological age, referred to as the DNA methylation clock. DNA methylation may also have value as a biomarker of healthy versus unhealthy aging and disease risk; in other words, a biological clock. Here we consider the relationship between the chronological and biological clocks, their underlying mechanisms, potential consequences, and their utility as biomarkers and as targets for intervention to promote healthy aging and longevity.
Collapse
|
164
|
Ciabattini A, Nardini C, Santoro F, Garagnani P, Franceschi C, Medaglini D. Vaccination in the elderly: The challenge of immune changes with aging. Semin Immunol 2019; 40:83-94. [PMID: 30501873 DOI: 10.1016/j.smim.2018.10.010] [Citation(s) in RCA: 272] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/13/2022]
Abstract
The unprecedented increase of life expectancy challenges society to protect the elderly from morbidity and mortality making vaccination a crucial mean to safeguard this population. Indeed, infectious diseases, such as influenza and pneumonia, are among the top killers of elderly people in the world. Elderly individuals are more prone to severe infections and less responsive to vaccination prevention, due to immunosenescence combined with the progressive increase of a proinflammatory status characteristic of the aging process (inflammaging). These factors are responsible for most age-related diseases and correlate with poor response to vaccination. Therefore, it is of utmost interest to deepen the knowledge regarding the role of inflammaging in vaccination responsiveness to support the development of effective vaccination strategies designed for elderly. In this review we analyse the impact of age-associated factors such as inflammaging, immunosenescence and immunobiography on immune response to vaccination in the elderly, and we consider systems biology approaches as a mean for integrating a multitude of data in order to rationally design vaccination approaches specifically tailored for the elderly.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Viale Bracci 16, 53100, Siena, Italy
| | - Christine Nardini
- Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, SE-171 77, Stockholm, Sweden; Personal Genomics S.r.l., Via Roveggia, 43B, 37134, Verona, Italy; CNR IAC "Mauro Picone", Via dei Taurini, 19, 00185, Roma, Italy
| | - Francesco Santoro
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Viale Bracci 16, 53100, Siena, Italy
| | - Paolo Garagnani
- Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, SE-171 77, Stockholm, Sweden; Interdepartmental Centre 'L. Galvani' (CIG), University of Bologna, Via G. Petroni 26, 40139, Bologna, Italy; Department of Experimental, Diagnostic and Specialty Medicine (DIMES) - University of Bologna,40139, Bologna, Italy
| | - Claudio Franceschi
- IRCCS, Institute of Neurological Sciences of Bologna, Via Altura 3, 40139, Bologna, Italy.
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Viale Bracci 16, 53100, Siena, Italy.
| |
Collapse
|
165
|
Søraas A, Matsuyama M, de Lima M, Wald D, Buechner J, Gedde-Dahl T, Søraas CL, Chen B, Ferrucci L, Dahl JA, Horvath S, Matsuyama S. Epigenetic age is a cell-intrinsic property in transplanted human hematopoietic cells. Aging Cell 2019; 18:e12897. [PMID: 30712319 PMCID: PMC6413751 DOI: 10.1111/acel.12897] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 10/16/2018] [Accepted: 11/03/2018] [Indexed: 12/21/2022] Open
Abstract
The age of tissues and cells can be accurately estimated by DNA methylation analysis. The multitissue DNA methylation (DNAm) age predictor combines the DNAm levels of 353 CpG dinucleotides to arrive at an age estimate referred to as DNAm age. Recent studies based on short‐term observations showed that the DNAm age of reconstituted blood following allogeneic hematopoietic stem cell transplantation (HSCT) reflects the age of the donor. However, it is not known whether the DNAm age of donor blood remains independent of the recipient's age over the long term. Importantly, long‐term studies including child recipients have the potential to clearly reveal whether DNAm age is cell‐intrinsic or whether it is modulated by extracellular cues in vivo. Here, we address this question by analyzing blood methylation data from HSCT donor and recipient pairs who greatly differed in chronological age (age differences between 1 and 49 years). We found that the DNAm age of the reconstituted blood was not influenced by the recipient's age, even 17 years after HSCT, in individuals without relapse of their hematologic disorder. However, the DNAm age of recipients with relapse of leukemia was unstable. These data are consistent with our previous findings concerning the abnormal DNAm age of cancer cells, and it can potentially be exploited to monitor the health of HSCT recipients. Our data demonstrate that transplanted human hematopoietic stem cells have an intrinsic DNAm age that is unaffected by the environment in a recipient of a different age.
Collapse
Affiliation(s)
- Arne Søraas
- Department of Microbiology; Oslo University Hospital; Oslo Norway
| | - Mieko Matsuyama
- Division of Hematology/Oncology, Department of Medicine, School of Medicine; Case Western Reserve University; Cleveland Ohio
| | - Marcos de Lima
- Division of Hematology/Oncology, Department of Medicine, School of Medicine; Case Western Reserve University; Cleveland Ohio
- Stem Cell Transplant Program, University Hospitals of Cleveland; Case Western Reserve University; Cleveland Ohio
| | - David Wald
- Department of Pathology; Case Western Reserve University; Cleveland Ohio
| | - Jochen Buechner
- Department of Pediatric Hematology and Oncology; Oslo University Hospital; Oslo Norway
| | - Tobias Gedde-Dahl
- Department of Hematology; Oslo University Hospital; Oslo Norway
- Institute of Clinical Medicine; University of Oslo; Oslo Norway
| | | | - Brian Chen
- National Institute of Aging (NIA); National Institute of Health; Bethesda Maryland
| | - Luigi Ferrucci
- National Institute of Aging (NIA); National Institute of Health; Bethesda Maryland
| | - John Arne Dahl
- Department of Microbiology; Oslo University Hospital; Oslo Norway
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine; University of California, Los Angeles; Los Angeles California
- Department of Biostatistics, Fielding School of Public Health; University of California, Los Angeles; Los Angeles California
| | - Shigemi Matsuyama
- Division of Hematology/Oncology, Department of Medicine, School of Medicine; Case Western Reserve University; Cleveland Ohio
- Case Comprehensive Cancer Center; Case Western Reserve University; Cleveland Ohio
| |
Collapse
|
166
|
Lung Cancer Screening, Towards a Multidimensional Approach: Why and How? Cancers (Basel) 2019; 11:cancers11020212. [PMID: 30759893 PMCID: PMC6406662 DOI: 10.3390/cancers11020212] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/06/2019] [Accepted: 02/06/2019] [Indexed: 12/19/2022] Open
Abstract
Early-stage treatment improves prognosis of lung cancer and two large randomized controlled trials have shown that early detection with low-dose computed tomography (LDCT) reduces mortality. Despite this, lung cancer screening (LCS) remains challenging. In the context of a global shortage of radiologists, the high rate of false-positive LDCT results in overloading of existing lung cancer clinics and multidisciplinary teams. Thus, to provide patients with earlier access to life-saving surgical interventions, there is an urgent need to improve LDCT-based LCS and especially to reduce the false-positive rate that plagues the current detection technology. In this context, LCS can be improved in three ways: (1) by refining selection criteria (risk factor assessment), (2) by using Computer Aided Diagnosis (CAD) to make it easier to interpret chest CTs, and (3) by using biological blood signatures for early cancer detection, to both spot the optimal target population and help classify lung nodules. These three main ways of improving LCS are discussed in this review.
Collapse
|
167
|
Epigenetic age acceleration is associated with allergy and asthma in children in Project Viva. J Allergy Clin Immunol 2019; 143:2263-2270.e14. [PMID: 30738172 DOI: 10.1016/j.jaci.2019.01.034] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Epigenetic clocks have been suggested to capture one feature of the complexity between aging and the epigenome. However, little is known about the epigenetic clock in childhood allergy and asthma. OBJECTIVE We sought to examine associations of DNA methylation age (DNAmAge) and epigenetic age acceleration with childhood allergy and asthma. METHODS We calculated DNAmAge and age acceleration at birth, early childhood, and midchildhood based on the IlluminaHumanMethylation450BeadChip in Project Viva. We evaluated epigenetic clock associations with allergy and asthma using covariate-adjusted linear and logistic regressions. We attempted to replicate our findings in the Genetics of Asthma in Costa Rica Study. RESULTS At midchildhood (mean age, 7.8 years) in Project Viva, DNAmAge and age acceleration were cross-sectionally associated with greater total serum IgE levels and greater odds of atopic sensitization. Every 1-year increase in intrinsic epigenetic age acceleration was associated with a 1.22 (95% CI, 1.07-1.39), 1.17 (95% CI, 1.03-1.34), and 1.29 (95% CI, 1.12-1.49) greater odds of atopic sensitization and environmental and food allergen sensitization. DNAmAge and extrinsic epigenetic age acceleration were also cross-sectionally associated with current asthma at midchildhood. DNAmAge and age acceleration at birth and early childhood were not associated with midchildhood allergy or asthma. The midchildhood association between age acceleration and atopic sensitization were replicated in an independent data set. CONCLUSIONS Because the epigenetic clock might reflect immune and developmental components of biological aging, our study suggests pathways through which molecular epigenetic mechanisms of immunity, development, and maturation can interact along the age axis and associate with childhood allergy and asthma by midchildhood.
Collapse
|
168
|
Ashapkin VV, Kutueva LI, Vanyushin BF. Epigenetic Clock: Just a Convenient Marker or an Active Driver of Aging? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1178:175-206. [PMID: 31493228 DOI: 10.1007/978-3-030-25650-0_10] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A global DNA hypomethylation and local changes in the methylation levels of specific DNA loci occur during aging in mammals. Global hypomethylation mainly affects highly methylated repeat sequences, such as transposable elements; it is an essentially stochastic process usually referred to as "epigenetic drift." Specific changes in DNA methylation affect various genome sequences and could be either hypomethylation or hypermethylation, but the prevailing tendencies are hypermethylation of promoter sequences associated with CpG islands and hypomethylation of CpG poor genes. Methylation levels of multiple CpG sites display a strong correlation to age common between individuals of the same species. Collectively, methylation of such CpG sites could be used as "epigenetic clocks" to predict biological age. Furthermore, the discrepancy between epigenetic and chronological ages could be predictive of all-cause mortality and multiple age-associated diseases. Random changes in DNA methylation (epigenetic drift) could also affect the aging phenotype, causing accidental changes in gene expression and increasing the transcriptional noise between cells of the same tissue. Both effects could become detrimental to tissue functioning and cause a gradual decline in organ function during aging. Strong evidence shows that epigenetic systems contribute to lifespan control in various organisms. Similar to other cell systems, the epigenome is prone to gradual degradation due to the genome damage, stressful agents and other aging factors. However, unlike mutations and many other hallmarks of aging, age-related epigenetic changes could be fully or partially reversed to a "young" state.
Collapse
Affiliation(s)
- Vasily V Ashapkin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia.
| | - Lyudmila I Kutueva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Boris F Vanyushin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
169
|
Stevenson AJ, McCartney DL, Harris SE, Taylor AM, Redmond P, Starr JM, Zhang Q, McRae AF, Wray NR, Spires-Jones TL, McColl BW, McIntosh AM, Deary IJ, Marioni RE. Trajectories of inflammatory biomarkers over the eighth decade and their associations with immune cell profiles and epigenetic ageing. Clin Epigenetics 2018; 10:159. [PMID: 30572949 PMCID: PMC6302523 DOI: 10.1186/s13148-018-0585-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/12/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Epigenetic age acceleration (an older methylation age compared to chronological age) correlates strongly with various age-related morbidities and mortality. Chronic systemic inflammation is thought to be a hallmark of ageing, but the relationship between an increased epigenetic age and this likely key phenotype of ageing has not yet been extensively investigated. METHODS We modelled the trajectories of the inflammatory biomarkers C-reactive protein (CRP; measured using both a high- and low-sensitivity assay) and interleukin-6 (IL-6) over the eighth decade in the Lothian Birth Cohort 1936. Using linear mixed models, we investigated the association between CRP and immune cell profiles imputed from the methylation data and examined the cross-sectional and longitudinal association between the inflammatory biomarkers and two measures of epigenetic age acceleration, derived from the Horvath and Hannum epigenetic clocks. RESULTS We found that low-sensitivity CRP declined, high-sensitivity CRP did not change, and IL-6 increased over time within the cohort. CRP levels inversely associated with CD8+T cells and CD4+T cells and positively associated with senescent CD8+T cells, plasmablasts and granulocytes. Cross-sectionally, the Hannum, but not the Horvath, measure of age acceleration was positively associated with each of the inflammatory biomarkers, including a restricted measure of CRP (≤ 10 mg/L) likely reflecting levels relevant to chronic inflammation. CONCLUSIONS We found a divergent relationship between inflammation and immune system parameters in older age. We additionally report the Hannum measure of epigenetic age acceleration associated with an elevated inflammatory profile cross-sectionally, but not longitudinally.
Collapse
Affiliation(s)
- Anna J. Stevenson
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Daniel L. McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Sarah E. Harris
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Adele M. Taylor
- Department for Psychology, University of Edinburgh, Edinburgh, UK
| | - Paul Redmond
- Department for Psychology, University of Edinburgh, Edinburgh, UK
| | - John M. Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - Qian Zhang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland Australia
| | - Allan F. McRae
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland Australia
| | - Naomi R. Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland Australia
| | - Tara L. Spires-Jones
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Barry W. McColl
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Andrew M. McIntosh
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Ian J. Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Department for Psychology, University of Edinburgh, Edinburgh, UK
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
170
|
Justice JN, Ferrucci L, Newman AB, Aroda VR, Bahnson JL, Divers J, Espeland MA, Marcovina S, Pollak MN, Kritchevsky SB, Barzilai N, Kuchel GA. A framework for selection of blood-based biomarkers for geroscience-guided clinical trials: report from the TAME Biomarkers Workgroup. GeroScience 2018; 40:419-436. [PMID: 30151729 PMCID: PMC6294728 DOI: 10.1007/s11357-018-0042-y] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/15/2018] [Indexed: 12/25/2022] Open
Abstract
Recent advances indicate that biological aging is a potentially modifiable driver of late-life function and chronic disease and have led to the development of geroscience-guided therapeutic trials such as TAME (Targeting Aging with MEtformin). TAME is a proposed randomized clinical trial using metformin to affect molecular aging pathways to slow the incidence of age-related multi-morbidity and functional decline. In trials focusing on clinical end-points (e.g., disease diagnosis or death), biomarkers help show that the intervention is affecting the underlying aging biology before sufficient clinical events have accumulated to test the study hypothesis. Since there is no standard set of biomarkers of aging for clinical trials, an expert panel was convened and comprehensive literature reviews conducted to identify 258 initial candidate biomarkers of aging and age-related disease. Next selection criteria were derived and applied to refine this set emphasizing: (1) measurement reliability and feasibility; (2) relevance to aging; (3) robust and consistent ability to predict all-cause mortality, clinical and functional outcomes; and (4) responsiveness to intervention. Application of these selection criteria to the current literature resulted in a short list of blood-based biomarkers proposed for TAME: IL-6, TNFα-receptor I or II, CRP, GDF15, insulin, IGF1, cystatin C, NT-proBNP, and hemoglobin A1c. The present report provides a conceptual framework for the selection of blood-based biomarkers for use in geroscience-guided clinical trials. This work also revealed the scarcity of well-vetted biomarkers for human studies that reflect underlying biologic aging hallmarks, and the need to leverage proposed trials for future biomarker discovery and validation.
Collapse
Affiliation(s)
- Jamie N Justice
- Internal Medicine Section on Gerontology and Geriatrics, and the Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA.
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Anne B Newman
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Vanita R Aroda
- Department of Medicine, Division of Diabetes, Endocrinology, and Hypertension Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Judy L Bahnson
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Jasmin Divers
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Mark A Espeland
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Santica Marcovina
- Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA, 98109, USA
| | - Michael N Pollak
- Department of Oncology, Jewish General Hospital, McGill University, Montreal, Quebec, H3T1E2, Canada
| | - Stephen B Kritchevsky
- Internal Medicine Section on Gerontology and Geriatrics, and the Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, 1 Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Nir Barzilai
- Department of Medicine, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| |
Collapse
|
171
|
Liu Z, Kuo PL, Horvath S, Crimmins E, Ferrucci L, Levine M. A new aging measure captures morbidity and mortality risk across diverse subpopulations from NHANES IV: A cohort study. PLoS Med 2018; 15:e1002718. [PMID: 30596641 PMCID: PMC6312200 DOI: 10.1371/journal.pmed.1002718] [Citation(s) in RCA: 309] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/20/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND A person's rate of aging has important implications for his/her risk of death and disease; thus, quantifying aging using observable characteristics has important applications for clinical, basic, and observational research. Based on routine clinical chemistry biomarkers, we previously developed a novel aging measure, Phenotypic Age, representing the expected age within the population that corresponds to a person's estimated mortality risk. The aim of this study was to assess its applicability for differentiating risk for a variety of health outcomes within diverse subpopulations that include healthy and unhealthy groups, distinct age groups, and persons with various race/ethnic, socioeconomic, and health behavior characteristics. METHODS AND FINDINGS Phenotypic Age was calculated based on a linear combination of chronological age and 9 multi-system clinical chemistry biomarkers in accordance with our previously established method. We also estimated Phenotypic Age Acceleration (PhenoAgeAccel), which represents Phenotypic Age after accounting for chronological age (i.e., whether a person appears older [positive value] or younger [negative value] than expected, physiologically). All analyses were conducted using NHANES IV (1999-2010, an independent sample from that originally used to develop the measure). Our analytic sample consisted of 11,432 adults aged 20-84 years and 185 oldest-old adults top-coded at age 85 years. We observed a total of 1,012 deaths, ascertained over 12.6 years of follow-up (based on National Death Index data through December 31, 2011). Proportional hazard models and receiver operating characteristic curves were used to evaluate all-cause and cause-specific mortality predictions. Overall, participants with more diseases had older Phenotypic Age. For instance, among young adults, those with 1 disease were 0.2 years older phenotypically than disease-free persons, and those with 2 or 3 diseases were about 0.6 years older phenotypically. After adjusting for chronological age and sex, Phenotypic Age was significantly associated with all-cause mortality and cause-specific mortality (with the exception of cerebrovascular disease mortality). Results for all-cause mortality were robust to stratifications by age, race/ethnicity, education, disease count, and health behaviors. Further, Phenotypic Age was associated with mortality among seemingly healthy participants-defined as those who reported being disease-free and who had normal BMI-as well as among oldest-old adults, even after adjustment for disease prevalence. The main limitation of this study was the lack of longitudinal data on Phenotypic Age and disease incidence. CONCLUSIONS In a nationally representative US adult population, Phenotypic Age was associated with mortality even after adjusting for chronological age. Overall, this association was robust across different stratifications, particularly by age, disease count, health behaviors, and cause of death. We also observed a strong association between Phenotypic Age and the disease count an individual had. These findings suggest that this new aging measure may serve as a useful tool to facilitate identification of at-risk individuals and evaluation of the efficacy of interventions, and may also facilitate investigation into potential biological mechanisms of aging. Nevertheless, further evaluation in other cohorts is needed.
Collapse
Affiliation(s)
- Zuyun Liu
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Pei-Lun Kuo
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, California, United States of America
| | - Eileen Crimmins
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Morgan Levine
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, United States of America
- Department of Epidemiology, Yale School of Public Health, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
172
|
Guarrera S, Viberti C, Cugliari G, Allione A, Casalone E, Betti M, Ferrante D, Aspesi A, Casadio C, Grosso F, Libener R, Piccolini E, Mirabelli D, Dianzani I, Magnani C, Matullo G. Peripheral Blood DNA Methylation as Potential Biomarker of Malignant Pleural Mesothelioma in Asbestos-Exposed Subjects. J Thorac Oncol 2018; 14:527-539. [PMID: 30408567 DOI: 10.1016/j.jtho.2018.10.163] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 10/02/2018] [Accepted: 10/27/2018] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Malignant pleural mesothelioma (MPM) is an aggressive tumor strongly associated with asbestos exposure. Patients are usually diagnosed when current treatments have limited benefits, highlighting the need for noninvasive early diagnostic tests to monitor asbestos-exposed people. METHODS We used a genome-wide methylation array to identify, in asbestos-exposed subjects, novel blood DNA methylation markers of MPM in 163 MPM cases and 137 cancer-free controls (82 MPM cases and 68 controls, training set; replication in 81 MPM cases and 69 controls, test set) sampled from the same areas. RESULTS Evidence of differential methylation between MPM cases and controls was found (more than 800 cytosine-guanine dinucleotide sites, false discovery rate p value (pfdr) < 0.05), mainly in immune system-related genes. Considering the top differentially methylated signals, seven single- cytosine-guanine dinucleotides and five genomic regions of coordinated methylation replicated with similar effect size in the test set (pfdr < 0.05). The top hypomethylated single-CpG (cases versus controls effect size less than -0.15, pfdr < 0.05 in both the training and test sets) was detected in FOXK1 (Forkhead-box K1) gene, an interactor of BAP1 which was found mutated in MPM tissue and as germline mutation in familial MPM. In the test set, comparison of receiver operating characteristic curves and the area under the curve (AUC) of two models, including or excluding methylation, showed a significant increase in case/control discrimination when considering DNA methylation together with asbestos exposure (AUC = 0.81 versus AUC = 0.89, DeLong's test p = 0.0013). CONCLUSIONS We identified signatures of differential methylation in DNA from whole blood between asbestos exposed MPM cases and controls. Our results provide the rationale to further investigate, in prospective studies, the potential use of blood DNA methylation profiles for the identification of early changes related to the MPM carcinogenic process.
Collapse
Affiliation(s)
- Simonetta Guarrera
- Italian Institute for Genomic Medicine, IIGM, Turin, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| | - Clara Viberti
- Italian Institute for Genomic Medicine, IIGM, Turin, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giovanni Cugliari
- Italian Institute for Genomic Medicine, IIGM, Turin, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alessandra Allione
- Italian Institute for Genomic Medicine, IIGM, Turin, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| | - Elisabetta Casalone
- Italian Institute for Genomic Medicine, IIGM, Turin, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| | - Marta Betti
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - Daniela Ferrante
- Medical Statistics and Cancer Epidemiology Unit, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy; Cancer Epidemiology Unit, CPO-Piemonte, Novara, Italy
| | - Anna Aspesi
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | | | - Federica Grosso
- Division of Medical Oncology, SS. Antonio e Biagio General Hospital, Alessandria, Italy
| | - Roberta Libener
- Pathology Unit, SS. Antonio e Biagio General Hospital, Alessandria, Italy
| | - Ezio Piccolini
- Pneumology Unit, Santo Spirito Hospital, Casale Monferrato (AL), Italy
| | - Dario Mirabelli
- Cancer Epidemiology Unit, Department of Medical Sciences, University of Turin, Turin, Italy; Cancer Epidemiology Unit, CPO Piemonte, Turin, Italy; Interdepartmental Center for Studies on Asbestos and Other Toxic Particulates "G. Scansetti," University of Turin, Turin, Italy
| | - Irma Dianzani
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy; Interdepartmental Center for Studies on Asbestos and Other Toxic Particulates "G. Scansetti," University of Turin, Turin, Italy
| | - Corrado Magnani
- Medical Statistics and Cancer Epidemiology Unit, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy; Cancer Epidemiology Unit, CPO-Piemonte, Novara, Italy; Interdepartmental Center for Studies on Asbestos and Other Toxic Particulates "G. Scansetti," University of Turin, Turin, Italy
| | - Giuseppe Matullo
- Italian Institute for Genomic Medicine, IIGM, Turin, Italy; Department of Medical Sciences, University of Turin, Turin, Italy; Interdepartmental Center for Studies on Asbestos and Other Toxic Particulates "G. Scansetti," University of Turin, Turin, Italy; Medical Genetics Unit, AOU Città della Salute e della Scienza, Turin, Italy.
| |
Collapse
|
173
|
Ward-Caviness CK, Huffman JE, Everett K, Germain M, van Dongen J, Hill WD, Jhun MA, Brody JA, Ghanbari M, Du L, Roetker NS, de Vries PS, Waldenberger M, Gieger C, Wolf P, Prokisch H, Koenig W, O'Donnell CJ, Levy D, Liu C, Truong V, Wells PS, Trégouët DA, Tang W, Morrison AC, Boerwinkle E, Wiggins KL, McKnight B, Guo X, Psaty BM, Sotoodenia N, Boomsma DI, Willemsen G, Ligthart L, Deary IJ, Zhao W, Ware EB, Kardia SLR, Van Meurs JBJ, Uitterlinden AG, Franco OH, Eriksson P, Franco-Cereceda A, Pankow JS, Johnson AD, Gagnon F, Morange PE, de Geus EJC, Starr JM, Smith JA, Dehghan A, Björck HM, Smith NL, Peters A. DNA methylation age is associated with an altered hemostatic profile in a multiethnic meta-analysis. Blood 2018; 132:1842-1850. [PMID: 30042098 PMCID: PMC6202911 DOI: 10.1182/blood-2018-02-831347] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 07/01/2018] [Indexed: 01/25/2023] Open
Abstract
Many hemostatic factors are associated with age and age-related diseases; however, much remains unknown about the biological mechanisms linking aging and hemostatic factors. DNA methylation is a novel means by which to assess epigenetic aging, which is a measure of age and the aging processes as determined by altered epigenetic states. We used a meta-analysis approach to examine the association between measures of epigenetic aging and hemostatic factors, as well as a clotting time measure. For fibrinogen, we performed European and African ancestry-specific meta-analyses which were then combined via a random effects meta-analysis. For all other measures we could not estimate ancestry-specific effects and used a single fixed effects meta-analysis. We found that 1-year higher extrinsic epigenetic age as compared with chronological age was associated with higher fibrinogen (0.004 g/L/y; 95% confidence interval, 0.001-0.007; P = .01) and plasminogen activator inhibitor 1 (PAI-1; 0.13 U/mL/y; 95% confidence interval, 0.07-0.20; P = 6.6 × 10-5) concentrations, as well as lower activated partial thromboplastin time, a measure of clotting time. We replicated PAI-1 associations using an independent cohort. To further elucidate potential functional mechanisms, we associated epigenetic aging with expression levels of the PAI-1 protein encoding gene (SERPINE1) and the 3 fibrinogen subunit-encoding genes (FGA, FGG, and FGB) in both peripheral blood and aorta intima-media samples. We observed associations between accelerated epigenetic aging and transcription of FGG in both tissues. Collectively, our results indicate that accelerated epigenetic aging is associated with a procoagulation hemostatic profile, and that epigenetic aging may regulate hemostasis in part via gene transcription.
Collapse
Affiliation(s)
- Cavin K Ward-Caviness
- Institute of Epidemiology II, Helmholtz Center of Munich, Neuherberg, Germany
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Chapel Hill, NC
| | - Jennifer E Huffman
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
- The Framingham Heart Study, Framingham, MA
- Center for Population Genomics, Boston VA Healthcare System, Jamaica Plain, MA
| | - Karl Everett
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Marine Germain
- Sorbonne Universités, UPMC University Paris 06, INSERM UMR_S 1166, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Jenny van Dongen
- Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - W David Hill
- Centre for Cognitive Ageing and Cognitive Epidemiology and
- Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Min A Jhun
- Department of Epidemiology, University of Michigan, Ann Arbor, MI
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Lei Du
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Nicholas S Roetker
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Melanie Waldenberger
- Institute of Epidemiology II, Helmholtz Center of Munich, Neuherberg, Germany
- Research Unit of Molecular Epidemiology and
| | | | - Petra Wolf
- Institue of Human Genetics, Helmholtz Center of Munich, Neuherberg, Germany
| | - Holger Prokisch
- Institue of Human Genetics, Helmholtz Center of Munich, Neuherberg, Germany
- Institute fur Humangenetik, Technische Univeritat Munchen, Munich, Germany
| | - Wolfgang Koenig
- Department of Internal Medicine II-Cardiology, University of Ulm Medical Center, Ulm, Germany
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Centre for Cardiovascular Research, Munich Heart Alliance, Munich, Germany
| | - Christopher J O'Donnell
- The Framingham Heart Study, Framingham, MA
- Cardiology Section Administration, Boston VA Healthcare System, West Roxbury, MA
| | - Daniel Levy
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
- The Framingham Heart Study, Framingham, MA
| | - Chunyu Liu
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
- The Framingham Heart Study, Framingham, MA
| | - Vinh Truong
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Philip S Wells
- Department of Medicine, University of Ottawa and Ottawa Hospital Research Institute, Ottawa, Canada
| | - David-Alexandre Trégouët
- Sorbonne Universités, UPMC University Paris 06, INSERM UMR_S 1166, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Weihong Tang
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Barbara McKnight
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Xiuqing Guo
- Department of Pediatrics, LABioMed at Harbor-UCLA Medical Center, Torrence, CA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
- Department of Epidemiology and
- Department of Health Services, University of Washington, Seattle, WA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA
| | - Nona Sotoodenia
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
- Division of Cardiology, University of Washington, Seattle, WA
| | - Dorret I Boomsma
- Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gonneke Willemsen
- Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lannie Ligthart
- Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology and
- Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI
| | - Erin B Ware
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
| | | | - Joyce B J Van Meurs
- Department of Internal Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Oscar H Franco
- Department of Epidemiology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Per Eriksson
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Anders Franco-Cereceda
- Cardiothoracic Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - James S Pankow
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Andrew D Johnson
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA
- The Framingham Heart Study, Framingham, MA
| | - France Gagnon
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Pierre-Emmanuel Morange
- Laboratory of Hematology, La Timone Hospital, Marseille, France
- INSERM UMR_S 1062, Nutrition Obesity and Risk of Thrombosis, Center for CardioVascular and Nutrition Research, Aix-Marseille University, Marseille, France
| | - Eco J C de Geus
- Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, VU Medical Center, Amsterdam, The Netherlands
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology and
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, United Kingdom; and
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Hanna M Björck
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Nicholas L Smith
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Epidemiology and
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA
- Seattle Epidemiologic Research and Information Center, Office of Research and Development, Department of Veterans Affairs, Seattle, WA
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Center of Munich, Neuherberg, Germany
| |
Collapse
|
174
|
Age-related DNA methylation and hemostatic factors. Blood 2018; 132:1736. [DOI: 10.1182/blood-2018-08-867028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
175
|
Ren JT, Wang MX, Su Y, Tang LY, Ren ZF. Decelerated DNA methylation age predicts poor prognosis of breast cancer. BMC Cancer 2018; 18:989. [PMID: 30333003 PMCID: PMC6191915 DOI: 10.1186/s12885-018-4884-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 10/01/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND DNA methylation (DNAm) age was found to be an indicator for all-cause mortality, cancer incidence, and longevity, but no study has involved in the associations of DNAm age with the prognosis of breast cancer. METHODS We retrieved information of 1076 breast cancer patients from Genomic Data Commons (GDC) data portal on March 30, 2017, including breast cancer DNAm profiling, demographic features, clinicopathological parameters, recurrence, and all-cause fatality. Horvath's method was applied to calculate the DNAm age. Cox proportional hazards regression models were used to test the associations between DNAm age of the cancerous tissues and the prognosis (recurrence of breast cancer and all-cause fatality) with or without adjusting for chronological age and clinicopathological parameters. RESULTS The DNAm age was markedly decelerated in the patients who were premenopausal, ER or PR negative, HER2-enriched or basal-like than their counterparts. In the first five-year follow-up dataset for survival, every ten-year increase in DNAm age was associated with a 15% decrease in fatality; subjects with DNAm age in the second (HR: 0.52; 95%CI: 0.29-0.92), the third (HR: 0.49; 95%CI: 0.27-0.87) and the fourth quartile (HR: 0.38; 95%CI: 0.20-0.72) had significant longer survival time than those in the first quartile. In the first five-year follow-up dataset for recurrence, every ten-year increase in DNAm age was associated with a 14% decrease of the recurrence; in the categorical analysis, a clear dose-response was shown (P for trend =0.02) and the fourth quartile was associated with a longer recurrence free survival (HR: 0.32; 95%CI: 0.14-0.74). In the full follow-up dataset, similar results were obtained. CONCLUSIONS DNAm age of breast cancer tissue, which associated with menopausal status and pathological features, was a strong independent predictor of the prognosis. It was suggested that the prognosis of breast cancer was related to intrinsic biological changes and specific molecular targets for treatment of breast cancer may be implicit.
Collapse
Affiliation(s)
- Jun-Ting Ren
- The School of Public Health, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China.,Mailman School of Public Health, Columbia University, New York, USA
| | - Mei-Xia Wang
- The School of Public Health, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China
| | - Yi Su
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Lu-Ying Tang
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ze-Fang Ren
- The School of Public Health, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
176
|
Marwitz S, Heinbockel L, Scheufele S, Kugler C, Reck M, Rabe KF, Perner S, Goldmann T, Ammerpohl O. Fountain of youth for squamous cell carcinomas? On the epigenetic age of non-small cell lung cancer and corresponding tumor-free lung tissues. Int J Cancer 2018; 143:3061-3070. [PMID: 29974462 PMCID: PMC6282761 DOI: 10.1002/ijc.31641] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/14/2018] [Accepted: 05/25/2018] [Indexed: 12/16/2022]
Abstract
Aging affects the core processes of almost every organism, and the functional decline at the cellular and tissue levels influences disease development. Recently, it was shown that the methylation of certain CpG dinucleotides correlates with chronological age and that this epigenetic clock can be applied to study aging‐related effects. We investigated these molecular age loci in non‐small cell lung cancer (NSCLC) tissues from patients with adenocarcinomas (AC) and squamous cell carcinomas (SQC) as well as in matched tumor‐free lung tissue. In both NSCLC subtypes, the calculated epigenetic age did not correlate with the chronological age. In particular, SQC exhibited rejuvenation compared to the corresponding normal lung tissue as well as with the chronological age of the donor. Moreover, the younger epigenetic pattern was associated with a trend toward stem cell‐like gene expression patterns. These findings show deep phenotypic differences between the tumor entities AC and SQC, which might be useful for novel therapeutic and diagnostic approaches. What's new? Chronological age is correlated with the methylation status of CpG sites in the genome, enabling the study of aging‐related phenomena. Here, investigation of molecular age loci in cells from patients with non‐small cell lung cancer (NSCLC) reveals remarkable differences in NSCLC cell epigenetic age compared to the host's chronological age. Adenocarcinomas showed a higher epigenetic age than squamous cell carcinomas (SQC). Reduced SQC epigenetic age was accompanied by increased expression of stem cell gene signatures, suggesting an increased abundance of stem cells in SQC. Elevated stem cell levels could have clinical implications, as stems cells often show therapeutic resistance.
Collapse
Affiliation(s)
- Sebastian Marwitz
- Pathology of the University Medical Center Schleswig‐Holstein (UKSH)Campus Luebeck and the Research Center Borstelsite BorstelGermany
- Airway Research Center North, Member of the German Center for Lung Research (DZL)GroßhansdorfGermany
| | - Lena Heinbockel
- Pathology of the University Medical Center Schleswig‐Holstein (UKSH)Campus Luebeck and the Research Center Borstelsite BorstelGermany
- Airway Research Center North, Member of the German Center for Lung Research (DZL)GroßhansdorfGermany
| | - Swetlana Scheufele
- Institute of Human Genetics, University Medical Center Schleswig‐Holstein (UKSH)Campus KielGermany
- Airway Research Center North, Member of the German Center for Lung Research (DZL)GroßhansdorfGermany
| | | | - Martin Reck
- OncologyLungenClinic GrosshansdorfGrosshansdorfGermany
- Airway Research Center North, Member of the German Center for Lung Research (DZL)GroßhansdorfGermany
| | - Klaus F. Rabe
- PneumologyLungenClinic GrosshansdorfGrosshansdorfGermany
- Airway Research Center North, Member of the German Center for Lung Research (DZL)GroßhansdorfGermany
| | - Sven Perner
- Pathology of the University Medical Center Schleswig‐Holstein (UKSH)Campus Luebeck and the Research Center Borstelsite BorstelGermany
| | - Torsten Goldmann
- Pathology of the University Medical Center Schleswig‐Holstein (UKSH)Campus Luebeck and the Research Center Borstelsite BorstelGermany
- Airway Research Center North, Member of the German Center for Lung Research (DZL)GroßhansdorfGermany
| | - Ole Ammerpohl
- Institute of Human Genetics, University Medical Center Ulm, UlmGermany
- Airway Research Center North, Member of the German Center for Lung Research (DZL)GroßhansdorfGermany
| |
Collapse
|
177
|
Tanaka T, Biancotto A, Moaddel R, Moore AZ, Gonzalez‐Freire M, Aon MA, Candia J, Zhang P, Cheung F, Fantoni G, Semba RD, Ferrucci L. Plasma proteomic signature of age in healthy humans. Aging Cell 2018; 17:e12799. [PMID: 29992704 PMCID: PMC6156492 DOI: 10.1111/acel.12799] [Citation(s) in RCA: 346] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/13/2018] [Accepted: 06/01/2018] [Indexed: 12/30/2022] Open
Abstract
To characterize the proteomic signature of chronological age, 1,301 proteins were measured in plasma using the SOMAscan assay (SomaLogic, Boulder, CO, USA) in a population of 240 healthy men and women, 22-93 years old, who were disease- and treatment-free and had no physical and cognitive impairment. Using a p ≤ 3.83 × 10-5 significance threshold, 197 proteins were positively associated, and 20 proteins were negatively associated with age. Growth differentiation factor 15 (GDF15) had the strongest, positive association with age (GDF15; 0.018 ± 0.001, p = 7.49 × 10-56 ). In our sample, GDF15 was not associated with other cardiovascular risk factors such as cholesterol or inflammatory markers. The functional pathways enriched in the 217 age-associated proteins included blood coagulation, chemokine and inflammatory pathways, axon guidance, peptidase activity, and apoptosis. Using elastic net regression models, we created a proteomic signature of age based on relative concentrations of 76 proteins that highly correlated with chronological age (r = 0.94). The generalizability of our findings needs replication in an independent cohort.
Collapse
Affiliation(s)
- Toshiko Tanaka
- Longitudinal Study SectionTranslational Gerontology BranchNIANIHBaltimoreMaryland
| | - Angelique Biancotto
- Trans‐NIH Center for Human Immunology, Autoimmunity, and InflammationNIHBethesdaMaryland
| | - Ruin Moaddel
- Laboratory of Clinical InvestigationNIANIHBaltimoreMaryland
| | - Ann Zenobia Moore
- Longitudinal Study SectionTranslational Gerontology BranchNIANIHBaltimoreMaryland
| | | | - Miguel A. Aon
- Laboratory of Cardiovascular ScienceNational Institute on AgingNational Institutes of HealthBaltimoreMaryland
| | - Julián Candia
- Trans‐NIH Center for Human Immunology, Autoimmunity, and InflammationNIHBethesdaMaryland
| | - Pingbo Zhang
- Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMaryland
| | - Foo Cheung
- Trans‐NIH Center for Human Immunology, Autoimmunity, and InflammationNIHBethesdaMaryland
| | - Giovanna Fantoni
- Trans‐NIH Center for Human Immunology, Autoimmunity, and InflammationNIHBethesdaMaryland
| | - Richard D. Semba
- Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMaryland
| | - Luigi Ferrucci
- Longitudinal Study SectionTranslational Gerontology BranchNIANIHBaltimoreMaryland
| |
Collapse
|
178
|
Mendelsohn AR, Larrick JW. Epigenetic Drift Is a Determinant of Mammalian Lifespan. Rejuvenation Res 2018; 20:430-436. [PMID: 28942711 DOI: 10.1089/rej.2017.2024] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The epigenome, which controls cell identity and function, is not maintained with 100% fidelity in somatic animal cells. Errors in the maintenance of the epigenome lead to epigenetic drift, an important hallmark of aging. Numerous studies have described DNA methylation clocks that correlate epigenetic drift with increasing age. The question of how significant a role epigenetic drift plays in creating the phenotypes associated with aging remains open. A recent study describes a new DNA methylation clock that can be slowed by caloric restriction (CR) in a way that correlates with the degree of lifespan and healthspan extension conferred by CR, suggesting that epigenetic drift itself is a determinant of mammalian lifespan. Genetic transplantation using genomic editing of DNA methylation homeostatic genes from long-lived to short-lived species is one way to potentially demonstrate a causative role for DNA methylation. Whether the DNA methylation clock be reset to youthful state, eliminating the effects of epigenetic drift without requiring a pluripotent cell intermediate is a critical question with profound implications for the development of aging therapeutics. Methods that transiently erase the DNA methylation pattern of somatic cells may be developed that reset this aging hallmark with potentially profound effects on lifespan, if DNA methylation-based epigenetic drift really plays a primary role in aging.
Collapse
Affiliation(s)
- Andrew R Mendelsohn
- 1 Regenerative Sciences Institute , Sunnyvale, California.,2 Panorama Research Institute , Sunnyvale, California
| | | |
Collapse
|
179
|
Hofstatter EW, Horvath S, Dalela D, Gupta P, Chagpar AB, Wali VB, Bossuyt V, Storniolo AM, Hatzis C, Patwardhan G, Von Wahlde MK, Butler M, Epstein L, Stavris K, Sturrock T, Au A, Kwei S, Pusztai L. Increased epigenetic age in normal breast tissue from luminal breast cancer patients. Clin Epigenetics 2018; 10:112. [PMID: 30157950 PMCID: PMC6114717 DOI: 10.1186/s13148-018-0534-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 07/23/2018] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Age is one of the most important risk factors for developing breast cancer. However, age-related changes in normal breast tissue that potentially lead to breast cancer are incompletely understood. Quantifying tissue-level DNA methylation can contribute to understanding these processes. We hypothesized that occurrence of breast cancer should be associated with an acceleration of epigenetic aging in normal breast tissue. RESULTS Ninety-six normal breast tissue samples were obtained from 88 subjects (breast cancer = 35 subjects/40 samples, unaffected = 53 subjects/53 samples). Normal tissue samples from breast cancer patients were obtained from distant non-tumor sites of primary mastectomy specimens, while samples from unaffected women were obtained from the Komen Tissue Bank (n = 25) and from non-cancer-related breast surgery specimens (n = 28). Patients were further stratified into four cohorts: age < 50 years with and without breast cancer and age ≥ 50 with and without breast cancer. The Illumina HumanMethylation450k BeadChip microarray was used to generate methylation profiles from extracted DNA samples. Data was analyzed using the "Epigenetic Clock," a published biomarker of aging based on a defined set of 353 CpGs in the human genome. The resulting age estimate, DNA methylation age, was related to chronological age and to breast cancer status. The DNAmAge of normal breast tissue was strongly correlated with chronological age (r = 0.712, p < 0.001). Compared to unaffected peers, breast cancer patients exhibited significant age acceleration in their normal breast tissue (p = 0.002). Multivariate analysis revealed that epigenetic age acceleration in the normal breast tissue of subjects with cancer remained significant after adjusting for clinical and demographic variables. Additionally, smoking was found to be positively correlated with epigenetic aging in normal breast tissue (p = 0.012). CONCLUSIONS Women with luminal breast cancer exhibit significant epigenetic age acceleration in normal adjacent breast tissue, which is consistent with an analogous finding in malignant breast tissue. Smoking is also associated with epigenetic age acceleration in normal breast tissue. Further studies are needed to determine whether epigenetic age acceleration in normal breast tissue is predictive of incident breast cancer and whether this mediates the risk of chronological age on breast cancer risk.
Collapse
Affiliation(s)
- Erin W. Hofstatter
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, 300 George Street, Suite 120, New Haven, CT 06511 USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095 USA
- Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Disha Dalela
- Department of Pharmacology, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06511 USA
| | - Piyush Gupta
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, 10065 USA
| | - Anees B. Chagpar
- Department of Surgery, Yale School of Medicine, 330 Cedar Street, New Haven, CT 06511 USA
| | - Vikram B. Wali
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, 300 George Street, Suite 120, New Haven, CT 06511 USA
| | - Veerle Bossuyt
- Department of Pathology, Yale School of Medicine, 330 Cedar Street, New Haven, CT 06511 USA
| | - Anna Maria Storniolo
- Department of Internal Medicine, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202 USA
| | - Christos Hatzis
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, 300 George Street, Suite 120, New Haven, CT 06511 USA
| | - Gauri Patwardhan
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, 300 George Street, Suite 120, New Haven, CT 06511 USA
| | - Marie-Kristin Von Wahlde
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, 300 George Street, Suite 120, New Haven, CT 06511 USA
- Department of Obstetrics and Gynecology, Münster University Hospital, Münster, Germany
| | - Meghan Butler
- Department of Pharmacology, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06511 USA
| | - Lianne Epstein
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, 300 George Street, Suite 120, New Haven, CT 06511 USA
| | - Karen Stavris
- Department of Pharmacology, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06511 USA
| | - Tracy Sturrock
- Department of Pharmacology, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06511 USA
| | - Alexander Au
- Department of Pharmacology, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06511 USA
- Department of Clinical Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Stephanie Kwei
- Department of Pharmacology, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06511 USA
| | - Lajos Pusztai
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, 300 George Street, Suite 120, New Haven, CT 06511 USA
| |
Collapse
|
180
|
Murphy SE, Park SL, Balbo S, Haiman CA, Hatsukami DK, Patel Y, Peterson LA, Stepanov I, Stram DO, Tretyakova N, Hecht SS, Le Marchand L. Tobacco biomarkers and genetic/epigenetic analysis to investigate ethnic/racial differences in lung cancer risk among smokers. NPJ Precis Oncol 2018; 2:17. [PMID: 30155522 PMCID: PMC6105591 DOI: 10.1038/s41698-018-0057-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/08/2018] [Accepted: 06/13/2018] [Indexed: 12/31/2022] Open
Abstract
The Multiethnic Cohort Study has demonstrated that African Americans and Native Hawaiians have a higher risk for lung cancer due to cigarette smoking than Whites while Latinos and Japanese Americans have a lower risk. These findings are consistent with other epidemiologic studies in the literature. In this review, we summarize tobacco carcinogen and toxicant biomarker studies and genetic analyses which partially explain these differences. As determined by measurement of total nicotine equivalents in urine, which account for about 85% of the nicotine dose, African Americans take up greater amounts of nicotine than Whites per cigarette while Japanese Americans take up less. There are corresponding differences in the uptake of tobacco smoke carcinogens such as tobacco-specific nitrosamines, polycyclic aromatic hydrocarbons, 1,3-butadiene, and other toxic volatiles. The lower nicotine uptake of Japanese Americans is clearly linked to the preponderance of low activity forms of the primary nicotine metabolizing enzyme CYP2A6 in this ethnic group, leading to more unchanged nicotine in the body and thus lower smoking intensity. But the relatively high risk of Native Hawaiians and the low risk of Latino smokers for lung cancer are not explained by these factors. The possible role of epigenetics in modifying lung cancer risk among smokers is also discussed here. The results of these published studies may lead to a better understanding of susceptibility factors for lung cancer in cigarette smokers thus potentially identifying biomarkers that can detect those individuals at highest risk so that preventive approaches can be initiated at an early stage of the lung cancer development process.
Collapse
Affiliation(s)
- Sharon E. Murphy
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455 USA
| | - Sungshim Lani Park
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089 USA
| | - Silvia Balbo
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455 USA
| | - Christopher A. Haiman
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089 USA
| | | | - Yesha Patel
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089 USA
| | - Lisa A. Peterson
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455 USA
| | - Irina Stepanov
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455 USA
| | - Daniel O. Stram
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089 USA
| | - Natalia Tretyakova
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455 USA
| | - Stephen S. Hecht
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455 USA
| | - Loïc Le Marchand
- Cancer Research Center of Hawaii, University of Hawaii, Honolulu, HI 96813 USA
| |
Collapse
|
181
|
Gale CR, Marioni RE, Harris SE, Starr JM, Deary IJ. DNA methylation and the epigenetic clock in relation to physical frailty in older people: the Lothian Birth Cohort 1936. Clin Epigenetics 2018; 10:101. [PMID: 30075802 PMCID: PMC6091041 DOI: 10.1186/s13148-018-0538-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/26/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The biological mechanisms underlying frailty in older people are poorly understood. There is some evidence to suggest that DNA methylation patterns may be altered in frail individuals. METHODS Participants were 791 people aged 70 years from the Lothian Birth Cohort 1936. DNA methylation was measured in whole blood. Biological age was estimated using two measures of DNA methylation-based age acceleration-extrinsic and intrinsic epigenetic age acceleration. We carried out an epigenome-wide association study of physical frailty, as defined by the Fried phenotype. Multinomial logistic regression was used to calculate relative risk ratios for being physically frail or pre-frail according to epigenetic age acceleration. RESULTS There was a single significant (P = 1.16 × 10-7) association in the epigenome-wide association study comparing frail versus not frail. The same CpG was not significant when comparing pre-frail versus not frail. Greater extrinsic epigenetic age acceleration was associated with an increased risk of being physically frail, but not of being pre-frail. For a year increase in extrinsic epigenetic age acceleration, age- and sex-adjusted relative risk ratios (95% CI) for being physically frail or pre-frail were 1.06 (1.02, 1.10) and 1.02 (1.00, 1.04), respectively. After further adjustment for smoking and chronic disease, the association with physical frailty remained significant. Intrinsic epigenetic age acceleration was not associated with physical frailty status. CONCLUSIONS People who are biologically older, as indexed by greater extrinsic epigenetic age acceleration, are more likely to be physically frail. Future research will need to investigate whether epigenetic age acceleration plays a causal role in the onset of physical frailty.
Collapse
Affiliation(s)
- Catharine R. Gale
- MRC Lifecourse Epidemiology Unit, Southampton General Hospital, University of Southampton, Southampton, SO16 6YD UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ UK
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ UK
| | - Riccardo E. Marioni
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ UK
- Medical Genetics Section, Centre for Genomics and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Sarah E. Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ UK
- Medical Genetics Section, Centre for Genomics and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - John M. Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, EH8 9JZ UK
| | - Ian J. Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ UK
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ UK
| |
Collapse
|
182
|
Nwanaji-Enwerem JC, Weisskopf MG, Baccarelli AA. Multi-tissue DNA methylation age: Molecular relationships and perspectives for advancing biomarker utility. Ageing Res Rev 2018; 45:15-23. [PMID: 29698722 PMCID: PMC6047923 DOI: 10.1016/j.arr.2018.04.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/29/2018] [Accepted: 04/18/2018] [Indexed: 12/31/2022]
Abstract
The multi-tissue DNA methylation estimator of chronological age (DNAm-age) has been associated with a wide range of exposures and health outcomes. Still, it is unclear how DNAm-age can have such broad relationships and how it can be best utilized as a biomarker. Understanding DNAm-age's molecular relationships is a promising approach to address this critical knowledge gap. In this review, we discuss the existing literature regarding DNAm-age's molecular relationships in six major categories: animal model systems, cancer processes, cellular aging processes, immune system processes, metabolic processes, and nucleic acid processes. We also present perspectives regarding the future of DNAm-age research, including the need to translate a greater number of ongoing research efforts to experimental and animal model systems.
Collapse
Affiliation(s)
- Jamaji C Nwanaji-Enwerem
- Department of Environmental Health, Harvard T.H. Chan School of Public Health and MD-PhD Program, Harvard Medical School, Boston, MA, USA.
| | - Marc G Weisskopf
- Department of Environmental Health and Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Columbia Mailman School of Public Health, New York, NY, USA
| |
Collapse
|
183
|
Horvath S, Stein DJ, Phillips N, Heany SJ, Kobor MS, Lin DTS, Myer L, Zar HJ, Levine AJ, Hoare J. Perinatally acquired HIV infection accelerates epigenetic aging in South African adolescents. AIDS 2018; 32:1465-1474. [PMID: 29746298 PMCID: PMC6026068 DOI: 10.1097/qad.0000000000001854] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Recent studies demonstrate that infection with the HIV-1 is associated with accelerated aging effects in adults according to a highly accurate epigenetic biomarker of aging known as epigenetic clock. However, it is not yet known whether epigenetic age acceleration occurs as early as adolescence in perinatally HIV-infected (PHIV+) youth. DESIGN Observational study of PHIV and HIV-uninfected adolescents enrolled in the Cape Town Adolescent Antiretroviral Cohort Study. METHODS The Illumina EPIC array was used to generate blood DNA methylation data from 204 PHIV and 44 age-matched, uninfected (HIV-) adolescents aged 9-12 years old. The epigenetic clock software and method was used to estimate two measures of epigenetic age acceleration. Each participant completed a comprehensive neuropsychological test battery upon enrollment to Cape Town Adolescent Antiretroviral Cohort. RESULTS HIV is associated with biologically older blood in PHIV+ adolescents according to both measures of epigenetic age acceleration. One of the measures, extrinsic epigenetic age acceleration, is negatively correlated with measures of cognitive functioning (executive functioning, working memory, processing speed). CONCLUSION Overall, our results indicate that epigenetic age acceleration in blood can be observed in PHIV+ adolescents and that these epigenetic changes accompany poorer cognitive functioning.
Collapse
Affiliation(s)
- Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine
- Department of Biostatistics, School of Public Health, University of California, Los Angeles, Los Angeles, California, USA
| | - Dan J Stein
- MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Mental Health, University of Cape Town, J-Block, Groote Schuur Hospital, Observatory, Cape Town, South Africa
| | - Nicole Phillips
- MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Mental Health, University of Cape Town, J-Block, Groote Schuur Hospital, Observatory, Cape Town, South Africa
| | - Sarah J Heany
- MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Mental Health, University of Cape Town, J-Block, Groote Schuur Hospital, Observatory, Cape Town, South Africa
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - David T S Lin
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Landon Myer
- Centre for Infectious Disease Epidemiology and Research
- Division of Epidemiology and Biostatistics, School of Public Health & Family Medicine, University of Cape Town
| | - Heather J Zar
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital
- SA-Medical Research Council Unit on Child and Adolescent Health, University of Cape Town, South Africa
| | - Andrew J Levine
- Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| | - Jacqueline Hoare
- MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Mental Health, University of Cape Town, J-Block, Groote Schuur Hospital, Observatory, Cape Town, South Africa
| |
Collapse
|
184
|
Khouja JN, Simpkin AJ, O'Keeffe LM, Wade KH, Houtepen LC, Relton CL, Suderman M, Howe LD. Epigenetic gestational age acceleration: a prospective cohort study investigating associations with familial, sociodemographic and birth characteristics. Clin Epigenetics 2018; 10:86. [PMID: 29983833 PMCID: PMC6020346 DOI: 10.1186/s13148-018-0520-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/14/2018] [Indexed: 11/20/2022] Open
Abstract
Background Gestational age at delivery is associated with health and social outcomes. Recently, cord blood DNA methylation data has been used to predict gestational age. The discrepancy between gestational age predicted from DNA methylation and determined by ultrasound or last menstrual period is known as gestational age acceleration. This study investigated associations of sex, socioeconomic status, parental behaviours and characteristics and birth outcomes with gestational age acceleration. Results Using data from the Avon Longitudinal Study of Parents and Children (n = 863), we found that pre-pregnancy maternal overweight and obesity were associated with greater gestational age acceleration (mean difference = 1.6 days, 95% CI 0.7 to 2.6, and 2.9 days, 95% CI 1.3 to 4.4, respectively, compared with a body mass index < 25 kg/m2, p < .001). There was evidence of an association between male sex and greater gestational age acceleration. Greater gestational age acceleration was associated with higher birthweight, birth length and head circumference of the child (mean differences per week higher gestational age acceleration: birthweight 0.1 kg, 95% CI 0.1 to 0.2, p < .001; birth length 0.4 cm, 95% CI 0.2 to 0.7, p < .001; head circumference 0.2 cm, 95% CI 0.1 to − 0.4, p < .001). There was evidence of an association between gestational age acceleration and mode of delivery (assisted versus unassisted delivery, odds ratio = 0.9 per week higher gestational age acceleration, 95% CI 0.7, 1.3 (p = .05); caesarean section versus unassisted delivery, odds ratio = 0.6, 95% CI 0.4 to 0.9 per week higher gestational age acceleration (p = .05)). There was no evidence of association for other parental and perinatal characteristics. Conclusions The associations of higher maternal body mass index and larger birth size with greater gestational age acceleration may imply that maternal overweight and obesity is associated with more rapid development of the fetus in utero. The implications of gestational age acceleration for postnatal health warrant further investigation. Electronic supplementary material The online version of this article (10.1186/s13148-018-0520-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jasmine N Khouja
- 1MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, England.,2Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England.,3School of Experimental Psychology at the University of Bristol, Bristol, England
| | - Andrew J Simpkin
- 1MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, England.,2Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England
| | - Linda M O'Keeffe
- 1MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, England.,2Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England
| | - Kaitlin H Wade
- 1MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, England.,2Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England
| | - Lotte C Houtepen
- 1MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, England.,2Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England
| | - Caroline L Relton
- 1MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, England.,2Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England
| | - Matthew Suderman
- 1MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, England.,2Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England
| | - Laura D Howe
- 1MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, England.,2Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England
| |
Collapse
|
185
|
Wolf EJ, Maniates H, Nugent N, Maihofer AX, Armstrong D, Ratanatharathorn A, Ashley-Koch AE, Garrett M, Kimbrel NA, Lori A, Aiello AE, Baker DG, Beckham JC, Boks MP, Galea S, Geuze E, Hauser MA, Kessler RC, Koenen KC, Miller MW, Ressler KJ, Risbrough V, Rutten BP, Stein MB, Ursano RJ, Vermetten E, Vinkers CH, Uddin M, Smith AK, Nievergelt CM, Logue MW. Traumatic stress and accelerated DNA methylation age: A meta-analysis. Psychoneuroendocrinology 2018; 92:123-134. [PMID: 29452766 PMCID: PMC5924645 DOI: 10.1016/j.psyneuen.2017.12.007] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/07/2017] [Accepted: 12/12/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Recent studies examining the association between posttraumatic stress disorder (PTSD) and accelerated aging, as defined by DNA methylation-based estimates of cellular age that exceed chronological age, have yielded mixed results. METHODS We conducted a meta-analysis of trauma exposure and PTSD diagnosis and symptom severity in association with accelerated DNA methylation age using data from 9 cohorts contributing to the Psychiatric Genomics Consortium PTSD Epigenetics Workgroup (combined N = 2186). Associations between demographic and cellular variables and accelerated DNA methylation age were also examined, as was the moderating influence of demographic variables. RESULTS Meta-analysis of regression coefficients from contributing cohorts revealed that childhood trauma exposure (when measured with the Childhood Trauma Questionnaire) and lifetime PTSD severity evidenced significant, albeit small, meta-analytic associations with accelerated DNA methylation age (ps = 0.028 and 0.016, respectively). Sex, CD4T cell proportions, and natural killer cell proportions were also significantly associated with accelerated DNA methylation age (all ps < 0.02). PTSD diagnosis and lifetime trauma exposure were not associated with advanced DNA methylation age. There was no evidence of moderation of the trauma or PTSD variables by demographic factors. CONCLUSIONS Results suggest that traumatic stress is associated with advanced epigenetic age and raise the possibility that cells integral to immune system maintenance and responsivity play a role in this. This study highlights the need for additional research into the biological mechanisms linking traumatic stress to accelerated DNA methylation age and the importance of furthering our understanding of the neurobiological and health consequences of PTSD.
Collapse
Affiliation(s)
- Erika J. Wolf
- National Center for PTSD at VA Boston Healthcare System,Department of Psychiatry, Boston University School of Medicine
| | | | - Nicole Nugent
- Bradley Hasbro Children’s Research Center, Rhode Island Hospital,Departments of Psychiatry and Human Behavior and Pediatrics, Brown Medical School
| | | | - Don Armstrong
- University of Illinois Urbana-Champaign, Carl R. Woese Institute for Genomic Biology
| | | | | | - Melanie Garrett
- Department of Psychiatry & Behavioral Sciences, Duke University Medical Center
| | - Nathan A. Kimbrel
- Department of Psychiatry & Behavioral Sciences, Duke University Medical Center,VA Mid-Atlantic, Mental Illness Research, Education, and Clinical Center,Durham VA Medical Center
| | - Adriana Lori
- Department of Psychiatry and Behavioral Sciences, Emory University
| | | | - Allison E. Aiello
- Department of Epidemiology, University of North Carolina at Chapel Hill Gillings School of Global Public Health
| | - Dewleen G. Baker
- University of California San Diego, Department of Psychiatry,Veterans Affairs San Diego Healthcare System,Veterans Affairs Center of Excellence for Stress and Mental Health
| | - Jean C. Beckham
- Department of Psychiatry & Behavioral Sciences, Duke University Medical Center,VA Mid-Atlantic, Mental Illness Research, Education, and Clinical Center,Durham VA Medical Center
| | - Marco P. Boks
- University Medical Center Utrecht, Brain Center Rudolf Magnus, Department of Psychiatry, Utrecht the Netherlands
| | | | - Elbert Geuze
- University Medical Center Utrecht, Brain Center Rudolf Magnus, Department of Psychiatry, Utrecht the Netherlands,Ministry of Defence, Military Mental Healthcare, Utrecht the Netherlands
| | - Michael A. Hauser
- Duke Molecular Physiology Institute, Duke University School of Medicine
| | | | - Karestan C. Koenen
- Harvard T.H. Chan School of Public Health, Department of Epidemiology,Massachusetts General Hospital, Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, and Department of Psychiatry
| | - Mark W. Miller
- National Center for PTSD at VA Boston Healthcare System,Department of Psychiatry, Boston University School of Medicine
| | - Kerry J. Ressler
- Department of Psychiatry, Harvard Medical School and McLean Hospital, Belmont, MA, USA
| | - Victoria Risbrough
- University of California San Diego, Department of Psychiatry,Veterans Affairs San Diego Healthcare System,Veterans Affairs Center of Excellence for Stress and Mental Health
| | - Bart P.F. Rutten
- School for Mental Health and Neuroscience and the European Graduate School of Neuroscience (EURON), Department of Psychiatry and Neuropsychology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Murray B. Stein
- University of California San Diego, Department of Psychiatry,Veterans Affairs San Diego Healthcare System,University of California San Diego, Department of Family Medicine and Public Health
| | - Robert J. Ursano
- Center for the Study of Traumatic Stress, Department of Psychiatry, Uniformed Services University of the Health Sciences
| | - Eric Vermetten
- University Medical Center Utrecht, Brain Center Rudolf Magnus, Department of Psychiatry, Utrecht the Netherlands,Ministry of Defence, Military Mental Healthcare, Utrecht the Netherlands,Arq Psychotrauma Expert Group
| | - Christiaan H. Vinkers
- University Medical Center Utrecht, Brain Center Rudolf Magnus, Department of Psychiatry, Utrecht the Netherlands
| | - Monica Uddin
- University of Illinois Urbana-Champaign, Carl R. Woese Institute for Genomic Biology,University of Illinois Urbana-Champaign, Department of Psychology
| | - Alicia K. Smith
- Department of Psychiatry and Behavioral Sciences, Emory University,Department of Gynecology and Obstetrics, Emory University
| | - Caroline M. Nievergelt
- University of California San Diego, Department of Psychiatry,Veterans Affairs San Diego Healthcare System,Veterans Affairs Center of Excellence for Stress and Mental Health
| | - Mark W. Logue
- National Center for PTSD at VA Boston Healthcare System,Department of Psychiatry, Boston University School of Medicine,Biomedical Genetics, Boston University School of Medicine
| |
Collapse
|
186
|
Inflammation and neutrophil immunosenescence in health and disease: Targeted treatments to improve clinical outcomes in the elderly. Exp Gerontol 2018; 105:70-77. [DOI: 10.1016/j.exger.2017.12.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 12/21/2017] [Accepted: 12/22/2017] [Indexed: 12/20/2022]
|
187
|
Gao Y, Widschwendter M, Teschendorff AE. DNA Methylation Patterns in Normal Tissue Correlate more Strongly with Breast Cancer Status than Copy-Number Variants. EBioMedicine 2018; 31:243-252. [PMID: 29735413 PMCID: PMC6013931 DOI: 10.1016/j.ebiom.2018.04.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/25/2018] [Accepted: 04/27/2018] [Indexed: 02/07/2023] Open
Abstract
Normal tissue at risk of neoplastic transformation is characterized by somatic mutations, copy-number variation and DNA methylation changes. It is unclear however, which type of alteration may be more informative of cancer risk. We analyzed genome-wide DNA methylation and copy-number calls from the same DNA assay in a cohort of healthy breast samples and age-matched normal samples collected adjacent to breast cancer. Using statistical methods to adjust for cell type heterogeneity, we show that DNA methylation changes can discriminate normal-adjacent from normal samples better than somatic copy-number variants. We validate this important finding in an independent dataset. These results suggest that DNA methylation alterations in the normal cell of origin may offer better cancer risk prediction and early detection markers than copy-number changes.
Collapse
Affiliation(s)
- Yang Gao
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Martin Widschwendter
- Department of Women's Cancer, University College London, 74 Huntley Street, London WC1E 6AU, United Kingdom
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; Department of Women's Cancer, University College London, 74 Huntley Street, London WC1E 6AU, United Kingdom; UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street, London WC1E 6BT, United Kingdom.
| |
Collapse
|
188
|
Levine ME, Lu AT, Quach A, Chen BH, Assimes TL, Bandinelli S, Hou L, Baccarelli AA, Stewart JD, Li Y, Whitsel EA, Wilson JG, Reiner AP, Aviv A, Lohman K, Liu Y, Ferrucci L, Horvath S. An epigenetic biomarker of aging for lifespan and healthspan. Aging (Albany NY) 2018; 10:573-591. [PMID: 29676998 PMCID: PMC5940111 DOI: 10.18632/aging.101414] [Citation(s) in RCA: 1761] [Impact Index Per Article: 251.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 04/08/2018] [Indexed: 04/08/2023]
Abstract
Identifying reliable biomarkers of aging is a major goal in geroscience. While the first generation of epigenetic biomarkers of aging were developed using chronological age as a surrogate for biological age, we hypothesized that incorporation of composite clinical measures of phenotypic age that capture differences in lifespan and healthspan may identify novel CpGs and facilitate the development of a more powerful epigenetic biomarker of aging. Using an innovative two-step process, we develop a new epigenetic biomarker of aging, DNAm PhenoAge, that strongly outperforms previous measures in regards to predictions for a variety of aging outcomes, including all-cause mortality, cancers, healthspan, physical functioning, and Alzheimer's disease. While this biomarker was developed using data from whole blood, it correlates strongly with age in every tissue and cell tested. Based on an in-depth transcriptional analysis in sorted cells, we find that increased epigenetic, relative to chronological age, is associated with increased activation of pro-inflammatory and interferon pathways, and decreased activation of transcriptional/translational machinery, DNA damage response, and mitochondrial signatures. Overall, this single epigenetic biomarker of aging is able to capture risks for an array of diverse outcomes across multiple tissues and cells, and provide insight into important pathways in aging.
Collapse
Affiliation(s)
- Morgan E. Levine
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ake T. Lu
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Austin Quach
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Brian H. Chen
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, USA. Baltimore, MD 21224, USA
| | | | | | - Lifang Hou
- Center for Population Epigenetics, Robert H. Lurie Comprehensive Cancer Center and Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Andrea A. Baccarelli
- Laboratory of Environmental Epigenetics, Departments of Environmental Health Sciences and Epidemiology, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - James D. Stewart
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yun Li
- Department of Genetics, Department of Biostatistics, Department of Computer Science, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eric A. Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Alex P Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Abraham Aviv
- Center of Human Development and Aging, New Jersey Medical School, Rutgers State University of New Jersey, Newark, NJ 07103, USA
| | - Kurt Lohman
- Center of Human Development and Aging, New Jersey Medical School, Rutgers State University of New Jersey, Newark, NJ 07103, USA
| | - Yongmei Liu
- Department of Epidemiology & Prevention, Division of Public Health Sciences, Wake Forrest School of Medicine, Winston-Salem, NC 27157, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, USA. Baltimore, MD 21224, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
189
|
Franceschi C, Garagnani P, Morsiani C, Conte M, Santoro A, Grignolio A, Monti D, Capri M, Salvioli S. The Continuum of Aging and Age-Related Diseases: Common Mechanisms but Different Rates. Front Med (Lausanne) 2018; 5:61. [PMID: 29662881 PMCID: PMC5890129 DOI: 10.3389/fmed.2018.00061] [Citation(s) in RCA: 540] [Impact Index Per Article: 77.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 02/20/2018] [Indexed: 12/11/2022] Open
Abstract
Geroscience, the new interdisciplinary field that aims to understand the relationship between aging and chronic age-related diseases (ARDs) and geriatric syndromes (GSs), is based on epidemiological evidence and experimental data that aging is the major risk factor for such pathologies and assumes that aging and ARDs/GSs share a common set of basic biological mechanisms. A consequence is that the primary target of medicine is to combat aging instead of any single ARD/GSs one by one, as favored by the fragmentation into hundreds of specialties and sub-specialties. If the same molecular and cellular mechanisms underpin both aging and ARDs/GSs, a major question emerges: which is the difference, if any, between aging and ARDs/GSs? The hypothesis that ARDs and GSs such as frailty can be conceptualized as accelerated aging will be discussed by analyzing in particular frailty, sarcopenia, chronic obstructive pulmonary disease, cancer, neurodegenerative diseases such as Alzheimer and Parkinson as well as Down syndrome as an example of progeroid syndrome. According to this integrated view, aging and ARDs/GSs become part of a continuum where precise boundaries do not exist and the two extremes are represented by centenarians, who largely avoided or postponed most ARDs/GSs and are characterized by decelerated aging, and patients who suffered one or more severe ARDs in their 60s, 70s, and 80s and show signs of accelerated aging, respectively. In between these two extremes, there is a continuum of intermediate trajectories representing a sort of gray area. Thus, clinically different, classical ARDs/GSs are, indeed, the result of peculiar combinations of alterations regarding the same, limited set of basic mechanisms shared with the aging process. Whether an individual will follow a trajectory of accelerated or decelerated aging will depend on his/her genetic background interacting lifelong with environmental and lifestyle factors. If ARDs and GSs are manifestations of accelerated aging, it is urgent to identify markers capable of distinguishing between biological and chronological age to identify subjects at higher risk of developing ARDs and GSs. To this aim, we propose the use of DNA methylation, N-glycans profiling, and gut microbiota composition to complement the available disease-specific markers.
Collapse
Affiliation(s)
- Claudio Franceschi
- Institute of Neurological Sciences, University of Bologna, Bellaria Hospital, Bologna, Italy
| | - Paolo Garagnani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden.,Applied Biomedical Research Center (CRBA), S. Orsola-Malpighi Polyclinic, Bologna, Italy.,CNR Institute of Molecular Genetics, Unit of Bologna, Bologna, Italy
| | - Cristina Morsiani
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Maria Conte
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Aurelia Santoro
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Interdepartmental Center "L. Galvani" (CIG), University of Bologna, Bologna, Italy
| | - Andrea Grignolio
- Unit and Museum of History of Medicine, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Miriam Capri
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Interdepartmental Center "L. Galvani" (CIG), University of Bologna, Bologna, Italy
| | - Stefano Salvioli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.,Interdepartmental Center "L. Galvani" (CIG), University of Bologna, Bologna, Italy
| |
Collapse
|
190
|
Zhang Y, Saum KU, Schöttker B, Holleczek B, Brenner H. Methylomic survival predictors, frailty, and mortality. Aging (Albany NY) 2018; 10:339-357. [PMID: 29514134 PMCID: PMC5892685 DOI: 10.18632/aging.101392] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 02/23/2018] [Indexed: 12/25/2022]
Abstract
Survival predictors are of potential use for informing on biological age and targeting prevention of aging-related morbidity. We assessed associations of 2 novel methylomic survival indicators, a methylation-based mortality risk score (MRscore) and the epigenetic clock-derived age acceleration (AA), with a well-known survival predictor, frailty index (FI), and compared the 3 indicators in mortality prediction. In a large population-based cohort with 14-year follow-up, we found both MRscore and AA to be independently associated with FI, but the association was much stronger for MRscore than for AA. Although all 3 indicators were individually associated with all-cause mortality, robust associations only persisted for MRscore and FI when simultaneously including the 3 indicators in regression models, with hazard ratios (95% CI) of 1.91 (1.63-2.22), 1.37 (1.25-1.51), and 1.05 (0.90-1.22), respectively, per standard deviation increase of MRscore, FI, and AA. Prediction error curves, Harrell's C-statistics, and time-dependent AUCs all showed higher predictive accuracy for MRscore than for FI and AA. These findings were validated in independent samples. Our study demonstrates the ability of the MRscore to strongly enhance survival prediction beyond established markers of biological age, such as FI and AA, and it thus bears potential of a surrogate endpoint for clinical research and intervention.
Collapse
Affiliation(s)
- Yan Zhang
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg D-69120, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg D-69120, Germany
| | - Kai-Uwe Saum
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg D-69120, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg D-69120, Germany
- Network Ageing Research, University of Heidelberg, Heidelberg 69115, Germany
| | | | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg D-69120, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg D-69120, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg D-69120, Germany
| |
Collapse
|
191
|
Li S, Wong EM, Dugué PA, McRae AF, Kim E, Joo JHE, Nguyen TL, Stone J, Dite GS, Armstrong NJ, Mather KA, Thalamuthu A, Wright MJ, Ames D, Milne RL, Craig JM, Saffery R, Montgomery GW, Song YM, Sung J, Spector TD, Sachdev PS, Giles GG, Southey MC, Hopper JL. Genome-wide average DNA methylation is determined in utero. Int J Epidemiol 2018. [PMID: 29518222 PMCID: PMC6005037 DOI: 10.1093/ije/dyy028] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Investigating the genetic and environmental causes of variation in genome-wide average DNA methylation (GWAM), a global methylation measure from the HumanMethylation450 array, might give a better understanding of genetic and environmental influences on methylation. METHODS We measured GWAM for 2299 individuals aged 0 to 90 years from seven twin and/or family studies. We estimated familial correlations, modelled correlations with cohabitation history and fitted variance components models for GWAM. RESULTS The correlation in GWAM for twin pairs was ∼0.8 at birth, decreased with age during adolescence and was constant at ∼0.4 throughout adulthood, with no evidence that twin pair correlations differed by zygosity. Non-twin first-degree relatives were correlated, from 0.17 [95% confidence interval (CI): 0.05-0.30] to 0.28 (95% CI: 0.08-0.48), except for middle-aged siblings (0.01, 95% CI: -0.10-0.12), and the correlation increased with time living together and decreased with time living apart. Spouse pairs were correlated in all studies, from 0.23 (95% CI: 0.3-0.43) to 0.31 (95% CI: 0.05-0.52), and the correlation increased with time living together. The variance explained by environmental factors shared by twins alone was 90% (95% CI: 74-95%) at birth, decreased in early life and plateaued at 28% (95% CI: 17-39%) in middle age and beyond. There was a cohabitation-related environmental component of variance. CONCLUSIONS GWAM is determined in utero by prenatal environmental factors, the effects of which persist throughout life. The variation of GWAM is also influenced by environmental factors shared by family members, as well as by individual-specific environmental factors.
Collapse
Affiliation(s)
- Shuai Li
- Centre for Epidemiology and Biostatistics
| | - Ee Ming Wong
- Genetic Epidemiology Laboratory, University of Melbourne, Parkville, VIC, Australia.,Precision Medicine, Monash University, Clayton, VIC, Australia
| | - Pierre-Antoine Dugué
- Centre for Epidemiology and Biostatistics.,Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Allan F McRae
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | - Eunae Kim
- Complex Disease and Genome Epidemiology Branch, Department of Public Health Science, School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Ji-Hoon Eric Joo
- Genetic Epidemiology Laboratory, University of Melbourne, Parkville, VIC, Australia.,Precision Medicine, Monash University, Clayton, VIC, Australia
| | | | - Jennifer Stone
- Centre for Genetic Origins of Health and Disease, Curtin University and the University of Western Australia, Perth, WA, Australia
| | | | | | - Karen A Mather
- Centre for Healthy Brain Ageing (CHeBA), University of New South Wales, Sydney, NSW, Australia
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing (CHeBA), University of New South Wales, Sydney, NSW, Australia
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | - David Ames
- National Ageing Research Institute and University of Melbourne Academic Unit for Psychiatry of Old Age, Parkville, VIC, Australia
| | - Roger L Milne
- Centre for Epidemiology and Biostatistics.,Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Jeffrey M Craig
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, VIC, Australia.,Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.,School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Richard Saffery
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, VIC, Australia.,Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Grant W Montgomery
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Yun-Mi Song
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Joohon Sung
- Complex Disease and Genome Epidemiology Branch, Department of Public Health Science, School of Public Health, Seoul National University, Seoul, Republic of Korea.,Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing (CHeBA), University of New South Wales, Sydney, NSW, Australia
| | - Graham G Giles
- Centre for Epidemiology and Biostatistics.,Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Melissa C Southey
- Genetic Epidemiology Laboratory, University of Melbourne, Parkville, VIC, Australia.,Precision Medicine, Monash University, Clayton, VIC, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics.,Complex Disease and Genome Epidemiology Branch, Department of Public Health Science, School of Public Health, Seoul National University, Seoul, Republic of Korea.,Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
192
|
Dugué PA, Bassett JK, Joo JE, Baglietto L, Jung CH, Wong EM, Fiorito G, Schmidt D, Makalic E, Li S, Moreno-Betancur M, Buchanan DD, Vineis P, English DR, Hopper JL, Severi G, Southey MC, Giles GG, Milne RL. Association of DNA Methylation-Based Biological Age With Health Risk Factors and Overall and Cause-Specific Mortality. Am J Epidemiol 2018; 187:529-538. [PMID: 29020168 DOI: 10.1093/aje/kwx291] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 07/26/2017] [Indexed: 11/13/2022] Open
Abstract
Measures of biological age based on blood DNA methylation, referred to as age acceleration (AA), have been developed. We examined whether AA was associated with health risk factors and overall and cause-specific mortality. At baseline (1990-1994), blood samples were drawn from 2,818 participants in the Melbourne Collaborative Cohort Study (Melbourne, Victoria, Australia). DNA methylation was determined using the Infinium HumanMethylation450 BeadChip array (Illumina Inc., San Diego, California). Mixed-effects models were used to examine the association of AA with health risk factors. Cox models were used to assess the association of AA with mortality. A total of 831 deaths were observed during a median 10.7 years of follow-up. Associations of AA were observed with male sex, Greek nationality (country of birth), smoking, obesity, diabetes, lower education, and meat intake. AA measures were associated with increased mortality, and this was only partly accounted for by known determinants of health (hazard ratios were attenuated by 20%-40%). Weak evidence of heterogeneity in the association was observed by sex (P = 0.06) and cause of death (P = 0.07) but not by other factors. DNA-methylation-based AA measures are associated with several major health risk factors, but these do not fully explain the association between AA and mortality. Future research should investigate what genetic and environmental factors determine AA.
Collapse
Affiliation(s)
- Pierre-Antoine Dugué
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
| | - Julie K Bassett
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia
| | - JiHoon E Joo
- Genetic Epidemiology Laboratory, Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
| | - Laura Baglietto
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
- Department of Clinical and Experimental Medicine, School of Medicine, University of Pisa, Pisa, Italy
| | - Chol-Hee Jung
- Melbourne Bioinformatics, University of Melbourne, Victoria, Australia
| | - Ee Ming Wong
- Genetic Epidemiology Laboratory, Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
| | | | - Daniel Schmidt
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
| | - Enes Makalic
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
| | - Shuai Li
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
| | - Margarita Moreno-Betancur
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
| | - Daniel D Buchanan
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
- Genetic Medicine and Familial Cancer Center, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Paolo Vineis
- Italian Institute for Genomic Medicine, Turin, Italy
- MRC-PHE Center for Environment and Health, Imperial College London, London, United Kingdom
| | - Dallas R English
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
| | - John L Hopper
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
| | - Gianluca Severi
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
- Centre de Recherche en Épidémiologie et Santé des Populations
- Italian Institute for Genomic Medicine, Turin, Italy
| | - Melissa C Southey
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
- Genetic Epidemiology Laboratory, Department of Pathology, University of Melbourne, Parkville, Victoria, Australia
| | - Graham G Giles
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
| | - Roger L Milne
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Center for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
193
|
Roetker NS, Pankow JS, Bressler J, Morrison AC, Boerwinkle E. Prospective Study of Epigenetic Age Acceleration and Incidence of Cardiovascular Disease Outcomes in the ARIC Study (Atherosclerosis Risk in Communities). CIRCULATION. GENOMIC AND PRECISION MEDICINE 2018; 11:e001937. [PMID: 29555670 PMCID: PMC5863591 DOI: 10.1161/circgen.117.001937] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/11/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND DNA methylation-based patterns of biological aging, known as epigenetic age acceleration, are predictive of all-cause mortality, but little is known about their association with cardiovascular disease (CVD). METHODS We estimated 2 versions of epigenetic age acceleration (Horvath and Hannum) using whole-blood samples from 2543 blacks. Linear and Cox proportional hazards regression, respectively, were used to assess the association of age acceleration with carotid intima-media thickness (cross-sectionally) and incident cardiovascular events, including CVD mortality, myocardial infarction, fatal coronary heart disease, peripheral arterial disease, and heart failure, during a median 21-year follow-up. All models were adjusted for chronological age and traditional CVD risk factors. RESULTS In comparison to chronological age, the 2 measures of epigenetic age acceleration were weaker, but independent, potential risk markers for subclinical atherosclerosis and most incident cardiovascular outcomes, including fatal coronary heart disease, peripheral arterial disease, and heart failure. For example, each 5-year increment of epigenetic age acceleration was associated with an average of 0.01 mm greater carotid intima-media thickness (each P≤0.01), and the hazard ratios (95% confidence intervals) of fatal coronary heart disease per 5-year increment in Horvath and Hannum age acceleration were 1.17 (1.02-1.33) and 1.22 (1.04-1.44), respectively. CONCLUSIONS In this sample of blacks, increased epigenetic age acceleration in whole blood was a potential risk marker for incident fatal coronary heart disease, peripheral arterial disease, and heart failure independently of chronological age and traditional CVD risk factors. DNA methylation-based measures of biological aging may help to identify new pathophysiological mechanisms contributing to the development of CVD.
Collapse
Affiliation(s)
- Nicholas S Roetker
- From the Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis (N.S.R., J.S.P.); Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston (J.B., A.C.M., E.B.); and Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX (E.B.).
| | - James S Pankow
- From the Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis (N.S.R., J.S.P.); Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston (J.B., A.C.M., E.B.); and Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX (E.B.)
| | - Jan Bressler
- From the Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis (N.S.R., J.S.P.); Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston (J.B., A.C.M., E.B.); and Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX (E.B.)
| | - Alanna C Morrison
- From the Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis (N.S.R., J.S.P.); Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston (J.B., A.C.M., E.B.); and Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX (E.B.)
| | - Eric Boerwinkle
- From the Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis (N.S.R., J.S.P.); Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston (J.B., A.C.M., E.B.); and Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX (E.B.)
| |
Collapse
|
194
|
Widschwendter M, Jones A, Evans I, Reisel D, Dillner J, Sundström K, Steyerberg EW, Vergouwe Y, Wegwarth O, Rebitschek FG, Siebert U, Sroczynski G, de Beaufort ID, Bolt I, Cibula D, Zikan M, Bjørge L, Colombo N, Harbeck N, Dudbridge F, Tasse AM, Knoppers BM, Joly Y, Teschendorff AE, Pashayan N. Epigenome-based cancer risk prediction: rationale, opportunities and challenges. Nat Rev Clin Oncol 2018; 15:292-309. [PMID: 29485132 DOI: 10.1038/nrclinonc.2018.30] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The incidence of cancer is continuing to rise and risk-tailored early diagnostic and/or primary prevention strategies are urgently required. The ideal risk-predictive test should: integrate the effects of both genetic and nongenetic factors and aim to capture these effects using an approach that is both biologically stable and technically reproducible; derive a score from easily accessible biological samples that acts as a surrogate for the organ in question; and enable the effectiveness of risk-reducing measures to be monitored. Substantial evidence has accumulated suggesting that the epigenome and, in particular, DNA methylation-based tests meet all of these requirements. However, the development and implementation of DNA methylation-based risk-prediction tests poses considerable challenges. In particular, the cell type specificity of DNA methylation and the extensive cellular heterogeneity of the easily accessible surrogate cells that might contain information relevant to less accessible tissues necessitates the use of novel methods in order to account for these confounding issues. Furthermore, the engagement of the scientific community with health-care professionals, policymakers and the public is required in order to identify and address the organizational, ethical, legal, social and economic challenges associated with the routine use of epigenetic testing.
Collapse
Affiliation(s)
- Martin Widschwendter
- Department of Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Allison Jones
- Department of Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Iona Evans
- Department of Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Daniel Reisel
- Department of Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Joakim Dillner
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Karin Sundström
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Ewout W Steyerberg
- Center for Medical Decision Sciences, Department of Public Health, Erasmus MC, Rotterdam, Netherlands.,Department of Biomedical Data Sciences, LUMC, Leiden, Netherlands
| | - Yvonne Vergouwe
- Center for Medical Decision Sciences, Department of Public Health, Erasmus MC, Rotterdam, Netherlands
| | - Odette Wegwarth
- Max Planck Institute for Human Development, Harding Center for Risk Literacy, Berlin, Germany.,Max Planck Institute for Human Development, Center for Adaptive Rationality, Berlin, Germany
| | - Felix G Rebitschek
- Max Planck Institute for Human Development, Harding Center for Risk Literacy, Berlin, Germany
| | - Uwe Siebert
- Institute of Public Health, Medical Decision Making and Health Technology Assessment, Department of Public Health, Health Services Research, and HTA, UMIT-University for Health Sciences, Medical Informatics and Technology, Hall in Tirol, Austria.,Harvard T. C. Chan School of Public Health, Center for Health Decision Science, Department of Health Policy and Management, Boston, MA, USA.,Oncotyrol: Center for Personalized Medicine, Innsbruck, Austria
| | - Gaby Sroczynski
- Institute of Public Health, Medical Decision Making and Health Technology Assessment, Department of Public Health, Health Services Research, and HTA, UMIT-University for Health Sciences, Medical Informatics and Technology, Hall in Tirol, Austria
| | - Inez D de Beaufort
- Department of Medical Ethics and Philosophy of Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Ineke Bolt
- Department of Medical Ethics and Philosophy of Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - David Cibula
- Department of Obstetrics and Gynaecology, First Medical Faculty of the Charles University and General Faculty Hospital, Prague, Czech Republic
| | - Michal Zikan
- Department of Obstetrics and Gynaecology, First Medical Faculty of the Charles University and General Faculty Hospital, Prague, Czech Republic
| | - Line Bjørge
- Department of Obstetrics and Gynecology, Haukeland University Hospital, and Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Nicoletta Colombo
- European Institute of Oncology and University Milan-Bicocca, Milan, Italy
| | - Nadia Harbeck
- Breast Center, Department of Gynaecology and Obstetrics, University of Munich (LMU), Munich, Germany
| | - Frank Dudbridge
- Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK.,Department of Health Sciences, University of Leicester, Leicester, UK
| | - Anne-Marie Tasse
- Public Population Project in Genomics and Society, McGill University and Genome Quebec Innovation Centre, Montreal, Canada
| | | | - Yann Joly
- Centre of Genomics and Policy, McGill University, Montreal, Canada
| | - Andrew E Teschendorff
- Department of Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Nora Pashayan
- Department of Applied Health Research, Institute of Epidemiology and Healthcare, University College London, UK
| | | |
Collapse
|
195
|
Maierhofer A, Flunkert J, Oshima J, Martin GM, Haaf T, Horvath S. Accelerated epigenetic aging in Werner syndrome. Aging (Albany NY) 2018; 9:1143-1152. [PMID: 28377537 PMCID: PMC5425119 DOI: 10.18632/aging.101217] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 04/23/2017] [Indexed: 11/25/2022]
Abstract
Individuals suffering from Werner syndrome (WS) exhibit many clinical signs of accelerated aging. While the underlying constitutional mutation leads to accelerated rates of DNA damage, it is not yet known whether WS is also associated with an increased epigenetic age according to a DNA methylation based biomarker of aging (the "Epigenetic Clock"). Using whole blood methylation data from 18 WS cases and 18 age matched controls, we find that WS is associated with increased extrinsic epigenetic age acceleration (p=0.0072) and intrinsic epigenetic age acceleration (p=0.04), the latter of which is independent of age-related changes in the composition of peripheral blood cells. A multivariate model analysis reveals that WS is associated with an increase in DNA methylation age (on average 6.4 years, p=0.011) even after adjusting for chronological age, gender, and blood cell counts. Further, WS might be associated with a reduction in naïve CD8+ T cells (p=0.025) according to imputed measures of blood cell counts. Overall, this study shows that WS is associated with an increased epigenetic age of blood cells which is independent of changes in blood cell composition. The extent to which this alteration is a cause or effect of WS disease phenotypes remains unknown.
Collapse
Affiliation(s)
- Anna Maierhofer
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| | - Julia Flunkert
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| | - Junko Oshima
- Department of Pathology, University of Washington, Seattle, WA 98105, USA.,Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - George M Martin
- Department of Pathology, University of Washington, Seattle, WA 98105, USA
| | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany.,Joint last authors
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA.,Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA.,Joint last authors
| |
Collapse
|
196
|
Binder AM, Corvalan C, Mericq V, Pereira A, Santos JL, Horvath S, Shepherd J, Michels KB. Faster ticking rate of the epigenetic clock is associated with faster pubertal development in girls. Epigenetics 2018; 13:85-94. [PMID: 29235933 DOI: 10.1080/15592294.2017.1414127] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Epigenetic age is an indicator of biological aging, capturing the impact of environmental and behavioral influences across time on cellular function. Deviance between epigenetic age and chronological age (AgeAccel) is a predictor of health. Pubertal timing has similarly been associated with cancer risk and mortality rate among females. We examined the association between AgeAccel and pubertal timing and adolescent breast composition in the longitudinal Growth and Obesity Cohort Study. AgeAccel was estimated in whole blood using the Horvath method at breast Tanner 2 (B2) and 4 (B4). Total breast volume, absolute fibro-glandular volume (FGV), and %FGV were evaluated at B4 using dual X-ray absorptiometry. The impact of AgeAccel (mean: 0; SD: 3.78) across puberty on the time to breast development (thelarche), menarche, and pubertal tempo (thelarche to menarche) was estimated using accelerated failure time models; generalized estimating equations were used to evaluate associations with breast density. A five-year increase in average adolescent AgeAccel was associated with a significant decrease in time to menarche [hazard ratio (HR): 1.37; 95% confidence interval (CI): 1.04, 1.80] adjusting for birth weight, maternal pre-pregnancy body mass index, maternal height, maternal education, B2 height, fat percentage, and cell composition. AgeAccel displayed a stronger inverse association with pubertal tempo (HR: 1.48; 95% CI: 1.10, 1.99). A five-year increase in AgeAccel was associated with 5% greater %FGV, adjusting for B4 percent body fat, and maternal traits (95% CI: 1.01, 1.10). Our study provides unique insight into the influence of AgeAccel on pubertal development in girls, which may have implications for adult health.
Collapse
Affiliation(s)
- Alexandra M Binder
- a Department of Epidemiology , Fielding School of Public Health, University of California , Los Angeles , 650 Charles E Young Drive South, Los Angeles , CA 90095 , USA
| | - Camila Corvalan
- b Institute of Nutrition and Food Technology , University of Chile , Av el Libano 5524, Santiago , Chile
| | - Verónica Mericq
- c Institute of Maternal and Child Research , University of Chile , Santa Rosa 1234, 2° piso, Santiago , Chile
| | - Ana Pereira
- b Institute of Nutrition and Food Technology , University of Chile , Av el Libano 5524, Santiago , Chile
| | - José Luis Santos
- d Department of Nutrition , Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile , Av Libertador Bernardo O'Higgins 340, Santiago , Chile
| | - Steve Horvath
- e Department of Biostatistics , School of Public Health, and Department of Human Genetics, Gonda Research Center , David Geffen School of Medicine, University of California, Los Angeles , 695 Charles E Young Drive South, Los Angeles , CA 90095 , USA
| | - John Shepherd
- f Department of Radiology and Biomedical Imaging , University of California, San Francisco , 400 Parnassus Avenue, San Francisco , CA 94117 , USA
| | - Karin B Michels
- a Department of Epidemiology , Fielding School of Public Health, University of California , Los Angeles , 650 Charles E Young Drive South, Los Angeles , CA 90095 , USA
| |
Collapse
|
197
|
Ward-Caviness CK, Nwanaji-Enwerem JC, Wolf K, Wahl S, Colicino E, Trevisi L, Kloog I, Just AC, Vokonas P, Cyrys J, Gieger C, Schwartz J, Baccarelli AA, Schneider A, Peters A. Long-term exposure to air pollution is associated with biological aging. Oncotarget 2018; 7:74510-74525. [PMID: 27793020 PMCID: PMC5342683 DOI: 10.18632/oncotarget.12903] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 10/13/2016] [Indexed: 11/28/2022] Open
Abstract
Long-term exposure to air pollution is associated with age-related diseases. We explored the association between accelerated biological aging and air pollution, a potential mechanism linking air pollution and health. We estimated long-term exposure to PM10, PM2.5, PM2.5 absorbance/black carbon (BC), and NOx via land-use regression models in individuals from the KORA F4 cohort. Accelerated biological aging was assessed using telomere length (TeloAA) and three epigenetic measures: DNA methylation age acceleration (DNAmAA), extrinsic epigenetic age acceleration (correlated with immune cell counts, EEAA), and intrinsic epigenetic age acceleration (independent of immune cell counts, IEAA). We also investigated sex-specific associations between air pollution and biological aging, given the published association between sex and aging measures. In KORA an interquartile range (0.97 μg/m3) increase in PM2.5 was associated with a 0.33 y increase in EEAA (CI = 0.01, 0.64; P = 0.04). BC and NOx (indicators or traffic exposure) were associated with DNAmAA and IEAA in women, while TeloAA was inversely associated with BC in men. We replicated this inverse BC-TeloAA association in the Normative Aging Study, a male cohort based in the USA. A multiple phenotype analysis in KORA F4 combining all aging measures showed that BC and PM10 were broadly associated with biological aging in men. Thus, we conclude that long-term exposure to air pollution is associated with biological aging measures, potentially in a sex-specific manner. However, many of the associations were relatively weak and further replication of overall and sex-specific associations is warranted.
Collapse
Affiliation(s)
- Cavin K Ward-Caviness
- Institute of Epidemiology II, Helmholtz Zentrum München, Neuherberg, Bavaria, Germany
| | | | - Kathrin Wolf
- Institute of Epidemiology II, Helmholtz Zentrum München, Neuherberg, Bavaria, Germany
| | - Simone Wahl
- Institute of Epidemiology II, Helmholtz Zentrum München, Neuherberg, Bavaria, Germany.,Research Unit Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Bavaria, Germany
| | - Elena Colicino
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Letizia Trevisi
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA
| | - Itai Kloog
- Department of Geography and Environmental Development, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Allan C Just
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pantel Vokonas
- VA Normative Aging Study, Veterans Affairs Boston Healthcare System and the Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Josef Cyrys
- Institute of Epidemiology II, Helmholtz Zentrum München, Neuherberg, Bavaria, Germany
| | - Christian Gieger
- Institute of Epidemiology II, Helmholtz Zentrum München, Neuherberg, Bavaria, Germany.,Research Unit Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Bavaria, Germany
| | - Joel Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Alexandra Schneider
- Institute of Epidemiology II, Helmholtz Zentrum München, Neuherberg, Bavaria, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München, Neuherberg, Bavaria, Germany
| |
Collapse
|
198
|
Declerck K, Vanden Berghe W. Back to the future: Epigenetic clock plasticity towards healthy aging. Mech Ageing Dev 2018; 174:18-29. [PMID: 29337038 DOI: 10.1016/j.mad.2018.01.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/22/2022]
Abstract
Aging is the most important risk factor for major human lifestyle diseases, including cancer, neurological and cardiometabolic disorders. Due to the complex interplay between genetics, lifestyle and environmental factors, some individuals seem to age faster than others, whereas centenarians seem to have a slower aging process. Therefore, a biochemical biomarker reflecting the relative biological age would be helpful to predict an individual's health status and aging disease risk. Although it is already known for years that cumulative epigenetic changes occur upon aging, DNA methylation patterns were only recently used to construct an epigenetic clock predictor for biological age, which is a measure of how well your body functions compared to your chronological age. Moreover, the epigenetic DNA methylation clock signature is increasingly applied as a biomarker to estimate aging disease susceptibility and mortality risk. Finally, the epigenetic clock signature could be used as a lifestyle management tool to monitor healthy aging, to evaluate preventive interventions against chronic aging disorders and to extend healthy lifespan. Dissecting the mechanism of the epigenetic aging clock will yield valuable insights into the aging process and how it can be manipulated to improve health span.
Collapse
Affiliation(s)
- Ken Declerck
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp (UA), Belgium
| | - Wim Vanden Berghe
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp (UA), Belgium.
| |
Collapse
|
199
|
Abstract
Cancer is largely an aging disease. Accelerated biological aging may be the strongest predictor of cancer and other chronic disease risks. In the absence of reliable and quantifiable biomarkers of aging to date, it has long been observed that tumorigenesis shares distinct epigenetic alterations with the aging process. Recently, epigenetic age estimates have been developed based on the availability of genome-wide DNA methylation profiles, by applying in the prediction formula the methylation level at a subset of highly predictive methylation sites, called epigenetic clock. These DNA methylation age estimates have produced remarkably strong correlations with chronological age, with a small deviation and high reproducibility across different age groups and study populations. Moreover, an increasing number of epidemiologic studies have demonstrated an independent association of DNA methylation age or the extent of acceleration with mortality and various aging-related conditions, even after accounting for differences in chronological age and other risk factors. Although epigenetic profiles are known to be tissue-specific, both target tissue- and multiple tissue-derived estimates appear to perform well to capture what is thought to be the cumulative epigenetic drift that represents a multifactorial degenerative process across tissues and organisms. Further refinement of the epigenetic age estimates is anticipated over time to accommodate a better technological coverage of the methylome and a better understanding of the biology underlying predictive regions. Epidemiologic principles will remain critical for the evaluation of research findings involving, for example, different study populations, design, follow-up time, and quality of covariate data. Overall, the epigenetic age estimates are an exciting development with useful implications for biomedical research of healthy aging and disease prevention and control.
Collapse
|
200
|
Dugué PA, Bassett JK, Joo JE, Jung CH, Ming Wong E, Moreno-Betancur M, Schmidt D, Makalic E, Li S, Severi G, Hodge AM, Buchanan DD, English DR, Hopper JL, Southey MC, Giles GG, Milne RL. DNA methylation-based biological aging and cancer risk and survival: Pooled analysis of seven prospective studies. Int J Cancer 2017; 142:1611-1619. [PMID: 29197076 DOI: 10.1002/ijc.31189] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/03/2017] [Accepted: 11/21/2017] [Indexed: 12/23/2022]
Abstract
The association between aging and cancer is complex. Recent studies have developed measures of biological aging based on DNA methylation and called them "age acceleration." We aimed to assess the associations of age acceleration with risk of and survival from seven common cancers. Seven case-control studies of DNA methylation and colorectal, gastric, kidney, lung, prostate and urothelial cancer and B-cell lymphoma nested in the Melbourne Collaborative Cohort Study were conducted. Cancer cases, vital status and cause of death were ascertained through linkage with cancer and death registries. Conditional logistic regression and Cox models were used to estimate odds ratios (OR) and hazard ratios (HR) and 95% confidence intervals (CI) for associations of five age acceleration measures derived from the Human Methylation 450 K Beadchip assay with cancer risk (N = 3,216 cases) and survival (N = 1,726 deaths), respectively. Epigenetic aging was associated with increased cancer risk, ranging from 4% to 9% per five-year age acceleration for the 5 measures considered. Heterogeneity by study was observed, with stronger associations for risk of kidney cancer and B-cell lymphoma. An associated increased risk of death following cancer diagnosis ranged from 2% to 6% per five-year age acceleration, with no evidence of heterogeneity by cancer site. Cancer risk and mortality were increased by 15-30% for the fourth versus first quartile of age acceleration. DNA methylation-based measures of biological aging are associated with increased cancer risk and shorter cancer survival, independently of major health risk factors.
Collapse
Affiliation(s)
- Pierre-Antoine Dugué
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Julie K Bassett
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia
| | - JiHoon E Joo
- Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Chol-Hee Jung
- Melbourne Bioinformatics, The University of Melbourne, Parkville, Victoria, Australia
| | - Ee Ming Wong
- Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Margarita Moreno-Betancur
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia.,Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Daniel Schmidt
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Enes Makalic
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Shuai Li
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Gianluca Severi
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia.,Centre de Recherche en Épidémiologie et Santé des Populations (CESP, Inserm U1018), Université Paris-Saclay, UPS, USQ, Gustave Roussy, Villejuif, France.,Human Genetics Foundation (HuGeF), Turin, Italy
| | - Allison M Hodge
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Daniel D Buchanan
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia.,Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia.,Genetic Medicine and Familial Cancer Centre, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Dallas R English
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Melissa C Southey
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia.,Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Graham G Giles
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Roger L Milne
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, Victoria, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|