1
|
Parrini I, Lucà F, Rao CM, Cacciatore S, Riccio C, Grimaldi M, Gulizia MM, Oliva F, Andreotti F. How to Manage Beta-Blockade in Older Heart Failure Patients: A Scoping Review. J Clin Med 2024; 13:2119. [PMID: 38610883 PMCID: PMC11012494 DOI: 10.3390/jcm13072119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Beta blockers (BBs) play a crucial role in enhancing the quality of life and extending the survival of patients with heart failure and reduced ejection fraction (HFrEF). Initiating the therapy at low doses and gradually titrating the dose upwards is recommended to ensure therapeutic efficacy while mitigating potential adverse effects. Vigilant monitoring for signs of drug intolerance is necessary, with dose adjustments as required. The management of older HF patients requires a case-centered approach, taking into account individual comorbidities, functional status, and frailty. Older adults, however, are often underrepresented in randomized clinical trials, leading to some uncertainty in management strategies as patients with HF in clinical practice are older than those enrolled in trials. The present article performs a scoping review of the past 25 years of published literature on BBs in older HF patients, focusing on age, outcomes, and tolerability. Twelve studies (eight randomized-controlled and four observational) encompassing 26,426 patients were reviewed. The results indicate that BBs represent a viable treatment for older HFrEF patients, offering benefits in symptom management, cardiac function, and overall outcomes. Their role in HF with preserved EF, however, remains uncertain. Further research is warranted to refine treatment strategies and address specific aspects in older adults, including proper dosing, therapeutic adherence, and tolerability.
Collapse
Affiliation(s)
- Iris Parrini
- Department of Cardiology, Mauriziano Hospital, Largo Filippo Turati, 62, 10128 Turin, Italy;
| | - Fabiana Lucà
- Cardiology Department, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, Via Melacrino 1, 89124 Reggio Calabria, Italy;
| | - Carmelo Massimiliano Rao
- Cardiology Department, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, Via Melacrino 1, 89124 Reggio Calabria, Italy;
| | - Stefano Cacciatore
- Department of Geriatrics, Orthopedics, and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy;
| | - Carmine Riccio
- Cardiovascular Department, Sant’Anna e San Sebastiano Hospital, Via Ferdinando Palasciano, 81100 Caserta, Italy;
| | - Massimo Grimaldi
- Department of Cardiology, General Regional Hospital “F. Miulli”, 70021 Bari, Italy;
| | | | - Fabrizio Oliva
- “A. De Gasperis” Cardiovascular Department, Division of Cardiology, ASST Grande Ospedale Metropolitano Niguarda, Piazza dell’Ospedale Maggiore 3, 20162 Milan, Italy;
| | - Felicita Andreotti
- Cardiovascular and Respiratory Sciences, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy;
| |
Collapse
|
2
|
Shen Y, Kioumourtzoglou MA, Wu H, Vokonas P, Spiro A, Navas-Acien A, Baccarelli AA, Gao F. Cohort Network: A Knowledge Graph toward Data Dissemination and Knowledge-Driven Discovery for Cohort Studies. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:8236-8244. [PMID: 37224396 PMCID: PMC10597774 DOI: 10.1021/acs.est.2c08174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Contemporary environmental health sciences draw on large-scale longitudinal studies to understand the impact of environmental exposures and behavior factors on the risk of disease and identify potential underlying mechanisms. In such studies, cohorts of individuals are assembled and followed up over time. Each cohort generates hundreds of publications, which are typically neither coherently organized nor summarized, hence limiting knowledge-driven dissemination. Hence, we propose a Cohort Network, a multilayer knowledge graph approach to extract exposures, outcomes, and their connections. We applied the Cohort Network on 121 peer-reviewed papers published over the past 10 years from the Veterans Affairs (VA) Normative Aging Study (NAS). The Cohort Network visualized connections between exposures and outcomes across different publications and identified key exposures and outcomes, such as air pollution, DNA methylation, and lung function. We demonstrated the utility of the Cohort Network for new hypothesis generation, e.g., identification of potential mediators of exposure-outcome associations. The Cohort Network can be used by investigators to summarize the cohort's research and facilitate knowledge-driven discovery and dissemination.
Collapse
Affiliation(s)
- Yike Shen
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Marianthi-Anna Kioumourtzoglou
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Haotian Wu
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Pantel Vokonas
- VA Normative Aging Study, VA Boston Healthcare System, Boston, Massachusetts 02130, United States
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts 02118, United States
| | - Avron Spiro
- VA Normative Aging Study, VA Boston Healthcare System, Boston, Massachusetts 02130, United States
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts 02118, United States
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts 02118, United States
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Feng Gao
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| |
Collapse
|
3
|
Uchehara B, Kwee LC, Regan J, Chatterjee R, Eckstrand J, Swope S, Gold G, Schaack T, Douglas P, Mettu P, Haddad F, Shore S, Hernandez A, Mahaffey KW, Pagidipati N, Shah SH. Accelerated Epigenetic Aging Is Associated With Multiple Cardiometabolic, Hematologic, and Renal Abnormalities: A Project Baseline Health Substudy. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:216-223. [PMID: 37039013 PMCID: PMC10330131 DOI: 10.1161/circgen.122.003772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 01/30/2023] [Indexed: 04/12/2023]
Abstract
BACKGROUND Epigenetic clocks estimate chronologic age using methylation levels at specific loci. We tested the hypothesis that accelerated epigenetic aging is associated with abnormal values in a range of clinical, imaging, and laboratory characteristics. METHODS The Project Baseline Health Study recruited 2502 participants, including 1661 with epigenetic age estimates from the Horvath pan-tissue clock. We classified individuals with extreme values as having epigenetic age acceleration (EAA) or epigenetic age deceleration. A subset of participants with longitudinal methylation profiling was categorized as accelerated versus nonaccelerated. Using principal components analysis, we created phenoclusters using 122 phenotypic variables and compared individuals with EAA versus epigenetic age deceleration, and at one year of follow-up, using logistic regression models adjusted for sex (false discovery rate [Q] <0.10); in secondary exploratory analyses, we tested individual clinical variables. RESULTS The EAA (n=188) and epigenetic age deceleration (n=195) groups were identified as having EAA estimates ≥5 years or ≤-5 years, respectively. In primary analyses, individuals with EAA had higher values for phenoclusters summarizing lung function and lipids, and lower values for a phenocluster representing physical function. In secondary analyses of individual variables, neutrophils, body mass index, and waist circumference were significantly higher in individuals with EAA (Q<0.10). No phenoclusters were significantly different between participants with accelerated (n=148) versus nonaccelerated (n=112) longitudinal aging. CONCLUSIONS We report multiple cardiometabolic, hematologic, and physical function features characterizing individuals with EAA. These highlight factors that may mediate the adverse effects of aging and identify potential targets for study of mitigation of these effects. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT03154346.
Collapse
Affiliation(s)
| | | | - Jessica Regan
- Division of General Internal Medicine, Dept of Medicine
| | | | | | - Sue Swope
- Stanford Center for Clinical Research, Dept of Medicine, Stanford University School of Medicine, Stanford
| | - Gary Gold
- Stanford Center for Clinical Research, Dept of Medicine, Stanford University School of Medicine, Stanford
| | - Terry Schaack
- California Health & Longevity Institute, Westlake Village
| | | | - Prithu Mettu
- Division of Retinal Ophthalmology, Dept of Ophthalmology
| | - Francois Haddad
- Stanford Center for Clinical Research, Dept of Medicine, Stanford University School of Medicine, Stanford
| | | | - Adrian Hernandez
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Kenneth W. Mahaffey
- Stanford Center for Clinical Research, Dept of Medicine, Stanford University School of Medicine, Stanford
| | | | - Svati H. Shah
- Duke Molecular Physiology Institute, Duke University
| |
Collapse
|
4
|
Kresovich JK, Sandler DP, Taylor JA. Methylation-Based Biological Age and Hypertension Prevalence and Incidence. Hypertension 2023; 80:1213-1222. [PMID: 36974720 PMCID: PMC10192055 DOI: 10.1161/hypertensionaha.122.20796] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/08/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND Hypertension is common in older individuals and is a major risk factor for cardiovascular disease. Blood DNA methylation profiles have been used to derive metrics of biological age that capture age-related physiological change, disease risk, and mortality. The relationships between hypertension and DNA methylation-based biological age metrics have yet to be carefully described. METHODS Among 4419 women enrolled in the prospective Sister Study cohort, DNA methylation data generated from whole blood samples collected at baseline were used to calculate 3 biological age metrics (PhenoAgeAccel, GrimAgeAccel, DunedinPACE). Women were classified as hypertensive at baseline if they had high blood pressure (systolic blood pressure ≥140 mm Hg or diastolic blood pressure ≥90 mm Hg) or reported current use of antihypertensive medication. New incident cases of hypertension during follow-up were identified via self-report on annual health questionnaires. RESULTS All 3 DNA methylation metrics of biological age were positively associated with prevalent hypertension at baseline (per 1-SD increase; PhenoAgeAccel, adjusted odds ratio, 1.16 [95% CI, 1.05-1.28]; GrimAgeAccel, adjusted odds ratio, 1.28 [95% CI, 1.14-1.45]; DunedinPACE, adjusted odds ratio, 1.16 [95% CI, 1.03-1.30]). Among 2610 women who were normotensive at baseline, women with higher biological age were more likely to be diagnosed with incident hypertension (per 1-SD increase; PhenoAgeAccel, adjusted hazard ratio, 1.09 [95% CI, 0.97-1.23]; GrimAgeAccel, adjusted hazard ratio, 1.16 [95% CI, 0.99-1.36]; DunedinPACE, adjusted hazard ratio, 1.16 [95% CI, 1.01-1.33]). CONCLUSIONS Methylation-based biological age metrics increase before a hypertension diagnosis and appear to remain elevated in the years after clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Jacob K Kresovich
- Departments of Cancer Epidemiology & Breast Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL (J.K.K.)
| | - Dale P Sandler
- Epidemiology Branch (D.P.S., J.A.T.), National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC
| | - Jack A Taylor
- Epigenetic and Stem Cell Biology Laboratory (J.A.T.), National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC
| |
Collapse
|
5
|
Tang B, Li X, Wang Y, Sjölander A, Johnell K, Thambisetty M, Ferrucci L, Reynolds CA, Finkel D, Jylhävä J, Pedersen NL, Hägg S. Longitudinal associations between use of antihypertensive, antidiabetic, and lipid-lowering medications and biological aging. GeroScience 2023:10.1007/s11357-023-00784-8. [PMID: 37032369 PMCID: PMC10400489 DOI: 10.1007/s11357-023-00784-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/26/2023] [Indexed: 04/11/2023] Open
Abstract
Aging is a major risk factor for many chronic diseases. This study aimed to examine the effects of antihypertensive, lipid-lowering, and antidiabetic drugs on biological aging. We included 672 participants and 2746 repeated measurements from the Swedish Adoption/Twin Study of Aging. Self-reported medicine uses were categorized into antidiabetic, antihypertensive, and lipid-lowering drugs. A total of 12 biomarkers for biological aging (BA biomarkers) were included as outcomes. Conditional generalized estimating equations were applied conditioning on individuals to estimate the drug effect on BA biomarker level within the same person when using or not using the drug. Chronological age, body mass index, smoking status, number of multiple medication uses, blood pressure, blood glucose level, and apoB/apoA ratio were adjusted for as covariates in the model. Overall, using antihypertensive drugs was associated with a decrease in one DNA-methylation age (PCGrimAge: beta = - 0.39, 95%CI = - 0.67 to - 0.12). When looking into drug subcategories, calcium channel blockers (CCBs) were associated with a decrease in several DNA-methylation ages (PCHorvathAge beta = - 1.28, 95%CI = - 2.34 to - 0.21; PCSkin&bloodAge beta = - 1.34, 95%CI = - 2.61 to - 0.07; PCPhenoAge beta = - 1.74, 95%CI = - 2.58 to - 0.89; PCGrimAge beta = - 0.57, 95%CI = - 0.96 to - 0.17) and in functional biological ages (functional age index beta = - 2.18, 95%CI = - 3.65 to - 0.71; frailty index beta = - 1.31, 95%CI = - 2.43 to - 0.18). However, the results within other drug subcategories were inconsistent. Calcium channel blockers may decrease biological aging captured by the BA biomarkers measured at epigenetic and functional level. Future studies are warranted to confirm these effects and understand the underlying biological mechanisms.
Collapse
Affiliation(s)
- Bowen Tang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Xia Li
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China
| | - Yunzhang Wang
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Solna, Sweden
| | - Arvid Sjölander
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Kristina Johnell
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Madhav Thambisetty
- Brain Aging and Behavior Section, National Institute on Aging, Baltimore, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, National Institute on Aging, Baltimore, USA
| | | | - Deborah Finkel
- Aging Research Network-Jönköping (ARN-J), School of Health and Welfare, Jönköping University, Jönköping, Sweden
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA
| | - Juulia Jylhävä
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
- Faculty of Social Sciences (Health Sciences) and Gerontology Research Center (GEREC), University of Tampere, Tampere, Finland
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
6
|
Yamazaki M, Yamada H, Munetsuna E, Maeda K, Ando Y, Mizuno G, Fujii R, Tsuboi Y, Ohashi K, Ishikawa H, Hashimoto S, Hamajima N, Suzuki K. DNA methylation level of the gene encoding thioredoxin-interacting protein in peripheral blood cells is associated with metabolic syndrome in the Japanese general population. Endocr J 2022; 69:319-326. [PMID: 34645728 DOI: 10.1507/endocrj.ej21-0339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Metabolic syndrome (MetS) is cluster of metabolic diseases, including abdominal obesity, hyperglycemia, high blood pressure, and dyslipidemia, that directly escalate the risk of type 2 diabetes, heart disease, and stroke. Thioredoxin-interacting protein (TXNIP) is a binding protein for thioredoxin, a molecule that is a key inhibitor of cellular oxidation, and thus regulates the cellular redox state. Epigenetic alteration of the TXNIP-encoding locus has been associated with components of MetS. In the present study, we sought to determine whether the level of TXNIP methylation in blood is associated with MetS in the general Japanese population. DNA was extracted from the peripheral blood cells of 37 subjects with and 392 subjects without MetS. The level of TXNIP methylation at cg19693031 was assessed by the bisulfite-pyrosequencing method. We observed that TXNIP methylation levels were lower in MetS subjects (median 74.9%, range 71.7-78.4%) than in non-MetS subjects (median 77.7%, range 74.4-80.5%; p = 0.0024). Calculation of the confounding factor-adjusted odds ratios (ORs) and 95% confidence intervals (CIs) for hypomethylation revealed that subjects with MetS exhibited significantly higher ORs for hypomethylation than did those without MetS (OR, 2.92; 95% CI, 1.33-6.62; p = 0.009). Our findings indicated that lower levels of TXNIP methylation are associated with MetS in the general Japanese population. Altered levels of DNA methylation in TXNIP at cg19693031 might play an important role in the pathogenesis of MetS.
Collapse
Affiliation(s)
- Mirai Yamazaki
- Department of Medical Technology, Kagawa Prefectural University of Health Sciences, Takamatsu 761-0123, Japan
| | - Hiroya Yamada
- Department of Hygiene, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Eiji Munetsuna
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Keisuke Maeda
- Department of Clinical Physiology, Fujita Health University School of Medical Sciences, Toyoake 470-1192, Japan
| | - Yoshitaka Ando
- Department of Biomedical and Analytical Sciences, Fujita Health University School of Medical Sciences, Toyoake 470-1192, Japan
| | - Genki Mizuno
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University Hospital, Toyoake 470-1192, Japan
| | - Ryosuke Fujii
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake 470-1192, Japan
| | - Yoshiki Tsuboi
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake 470-1192, Japan
| | - Koji Ohashi
- Department of Biomedical and Analytical Sciences, Fujita Health University School of Medical Sciences, Toyoake 470-1192, Japan
| | - Hiroaki Ishikawa
- Department of Biomedical and Analytical Sciences, Fujita Health University School of Medical Sciences, Toyoake 470-1192, Japan
| | - Shuji Hashimoto
- Department of Hygiene, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Nobuyuki Hamajima
- Department of Healthcare Administration, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Koji Suzuki
- Department of Preventive Medical Sciences, Fujita Health University School of Medical Sciences, Toyoake 470-1192, Japan
| |
Collapse
|
7
|
Malecki KMC, Andersen JK, Geller AM, Harry GJ, Jackson CL, James KA, Miller GW, Ottinger MA. Integrating Environment and Aging Research: Opportunities for Synergy and Acceleration. Front Aging Neurosci 2022; 14:824921. [PMID: 35264945 PMCID: PMC8901047 DOI: 10.3389/fnagi.2022.824921] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/12/2022] [Indexed: 12/25/2022] Open
Abstract
Despite significant overlaps in mission, the fields of environmental health sciences and aging biology are just beginning to intersect. It is increasingly clear that genetics alone does not predict an individual’s neurological aging and sensitivity to disease. Accordingly, aging neuroscience is a growing area of mutual interest within environmental health sciences. The impetus for this review came from a workshop hosted by the National Academies of Sciences, Engineering, and Medicine in June of 2020, which focused on integrating the science of aging and environmental health research. It is critical to bridge disciplines with multidisciplinary collaborations across toxicology, comparative biology, epidemiology to understand the impacts of environmental toxicant exposures and age-related outcomes. This scoping review aims to highlight overlaps and gaps in existing knowledge and identify essential research initiatives. It begins with an overview of aging biology and biomarkers, followed by examples of synergy with environmental health sciences. New areas for synergistic research and policy development are also discussed. Technological advances including next-generation sequencing and other-omics tools now offer new opportunities, including exposomic research, to integrate aging biomarkers into environmental health assessments and bridge disciplinary gaps. This is necessary to advance a more complete mechanistic understanding of how life-time exposures to toxicants and other physical and social stressors alter biological aging. New cumulative risk frameworks in environmental health sciences acknowledge that exposures and other external stressors can accumulate across the life course and the advancement of new biomarkers of exposure and response grounded in aging biology can support increased understanding of population vulnerability. Identifying the role of environmental stressors, broadly defined, on aging biology and neuroscience can similarly advance opportunities for intervention and translational research. Several areas of growing research interest include expanding exposomics and use of multi-omics, the microbiome as a mediator of environmental stressors, toxicant mixtures and neurobiology, and the role of structural and historical marginalization and racism in shaping persistent disparities in population aging and outcomes. Integrated foundational and translational aging biology research in environmental health sciences is needed to improve policy, reduce disparities, and enhance the quality of life for older individuals.
Collapse
Affiliation(s)
- Kristen M. C. Malecki
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- *Correspondence: Kristen M. C. Malecki,
| | | | - Andrew M. Geller
- United States Environmental Protection Agency, Office of Research and Development, Durham, NC, United States
| | - G. Jean Harry
- Division of National Toxicology Program, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Chandra L. Jackson
- Division of Intramural Research, Department of Health and Human Services, Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, United States
- Department of Health and Human Services, National Institute on Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, United States
| | - Katherine A. James
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado Denver, Denver, CO, United States
| | - Gary W. Miller
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, United States
| | - Mary Ann Ottinger
- Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| |
Collapse
|
8
|
Belsky DW, Caspi A, Corcoran DL, Sugden K, Poulton R, Arseneault L, Baccarelli A, Chamarti K, Gao X, Hannon E, Harrington HL, Houts R, Kothari M, Kwon D, Mill J, Schwartz J, Vokonas P, Wang C, Williams BS, Moffitt TE. DunedinPACE, a DNA methylation biomarker of the pace of aging. eLife 2022; 11:e73420. [PMID: 35029144 PMCID: PMC8853656 DOI: 10.7554/elife.73420] [Citation(s) in RCA: 408] [Impact Index Per Article: 136.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/13/2021] [Indexed: 01/09/2023] Open
Abstract
Background Measures to quantify changes in the pace of biological aging in response to intervention are needed to evaluate geroprotective interventions for humans. Previously, we showed that quantification of the pace of biological aging from a DNA-methylation blood test was possible (Belsky et al., 2020). Here, we report a next-generation DNA-methylation biomarker of Pace of Aging, DunedinPACE (for Pace of Aging Calculated from the Epigenome). Methods We used data from the Dunedin Study 1972-1973 birth cohort tracking within-individual decline in 19 indicators of organ-system integrity across four time points spanning two decades to model Pace of Aging. We distilled this two-decade Pace of Aging into a single-time-point DNA-methylation blood-test using elastic-net regression and a DNA-methylation dataset restricted to exclude probes with low test-retest reliability. We evaluated the resulting measure, named DunedinPACE, in five additional datasets. Results DunedinPACE showed high test-retest reliability, was associated with morbidity, disability, and mortality, and indicated faster aging in young adults with childhood adversity. DunedinPACE effect-sizes were similar to GrimAge Clock effect-sizes. In analysis of incident morbidity, disability, and mortality, DunedinPACE and added incremental prediction beyond GrimAge. Conclusions DunedinPACE is a novel blood biomarker of the pace of aging for gerontology and geroscience. Funding This research was supported by US-National Institute on Aging grants AG032282, AG061378, AG066887, and UK Medical Research Council grant MR/P005918/1.
Collapse
Affiliation(s)
- Daniel W Belsky
- Department of Epidemiology & Butler Columbia Aging Center, Columbia UniversityNew YorkUnited States
| | - Avshalom Caspi
- Center for Genomic and Computational Biology, Duke UniversityDurhamUnited States
| | - David L Corcoran
- Center for Genomic and Computational Biology, Duke UniversityDurhamUnited States
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Richie Poulton
- Department of Psychology, University of OtagoOtagoNew Zealand
| | - Louise Arseneault
- Social, Genetic, and Developmental Psychiatry Centre, King's College LondonLondonUnited Kingdom
| | - Andrea Baccarelli
- Department of Environmental Health Sciences, Columbia UniversityNew YorkUnited States
| | - Kartik Chamarti
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Xu Gao
- Department of Occupational and Environmental Health, Peking UniversityBeijingChina
| | - Eilis Hannon
- Complex Disease Epigenetics Group, University of ExeterExeterUnited Kingdom
| | - Hona Lee Harrington
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Renate Houts
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Meeraj Kothari
- Robert N Butler Columbia Aging Center, Columbia UniversityBrooklynUnited States
| | - Dayoon Kwon
- Robert N Butler Columbia Aging Center, Columbia UniversityNew YorkUnited States
| | - Jonathan Mill
- Complex Disease Epigenetics Group, University of ExeterExeterUnited Kingdom
| | - Joel Schwartz
- Department of Environmental Health Sciences, Harvard TH Chan School of Public Health, Harvard UniversityBostonUnited States
| | - Pantel Vokonas
- Department of Medicine, VA Boston Healthcare SystemBostonUnited States
| | - Cuicui Wang
- Department of Environmental Health Sciences, Harvard TH Chan School of Public Health, Harvard UniversityBostonUnited States
| | - Benjamin S Williams
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Terrie E Moffitt
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| |
Collapse
|
9
|
Oblak L, van der Zaag J, Higgins-Chen AT, Levine ME, Boks MP. A systematic review of biological, social and environmental factors associated with epigenetic clock acceleration. Ageing Res Rev 2021; 69:101348. [PMID: 33930583 DOI: 10.1016/j.arr.2021.101348] [Citation(s) in RCA: 290] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 04/01/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
Aging involves a diverse set of biological changes accumulating over time that leads to increased risk of morbidity and mortality. Epigenetic clocks are now widely used to quantify biological aging, in order to investigate determinants that modify the rate of aging and to predict age-related outcomes. Numerous biological, social and environmental factors have been investigated for their relationship to epigenetic clock acceleration and deceleration. The aim of this review was to synthesize general trends concerning the associations between human epigenetic clocks and these investigated factors. We conducted a systematic review of all available literature and included 156 publications across 4 resource databases. We compiled a list of all presently existing blood-based epigenetic clocks. Subsequently, we created an extensive dataset of over 1300 study findings in which epigenetic clocks were utilized in blood tissue of human subjects to assess the relationship between these clocks and numeral environmental exposures and human traits. Statistical analysis was possible on 57 such relationships, measured across 4 different epigenetic clocks (Hannum, Horvath, Levine and GrimAge). We found that the Horvath, Hannum, Levine and GrimAge epigenetic clocks tend to agree in direction of effects, but vary in size. Body mass index, HIV infection, and male sex were significantly associated with acceleration of one or more epigenetic clocks. Acceleration of epigenetic clocks was also significantly related to mortality, cardiovascular disease, cancer and diabetes. Our findings provide a graphical and numerical synopsis of the past decade of epigenetic age estimation research and indicate areas where further attention could be focused in the coming years.
Collapse
|
10
|
Xu H, Li S, Liu YS. Roles and Mechanisms of DNA Methylation in Vascular Aging and Related Diseases. Front Cell Dev Biol 2021; 9:699374. [PMID: 34262910 PMCID: PMC8273304 DOI: 10.3389/fcell.2021.699374] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/07/2021] [Indexed: 12/20/2022] Open
Abstract
Vascular aging is a pivotal risk factor promoting vascular dysfunction, the development and progression of vascular aging-related diseases. The structure and function of endothelial cells (ECs), vascular smooth muscle cells (VSMCs), fibroblasts, and macrophages are disrupted during the aging process, causing vascular cell senescence as well as vascular dysfunction. DNA methylation, an epigenetic mechanism, involves the alteration of gene transcription without changing the DNA sequence. It is a dynamically reversible process modulated by methyltransferases and demethyltransferases. Emerging evidence reveals that DNA methylation is implicated in the vascular aging process and plays a central role in regulating vascular aging-related diseases. In this review, we seek to clarify the mechanisms of DNA methylation in modulating ECs, VSMCs, fibroblasts, and macrophages functions and primarily focus on the connection between DNA methylation and vascular aging-related diseases. Therefore, we represent many vascular aging-related genes which are modulated by DNA methylation. Besides, we concentrate on the potential clinical application of DNA methylation to serve as a reliable diagnostic tool and DNA methylation-based therapeutic drugs for vascular aging-related diseases.
Collapse
Affiliation(s)
- Hui Xu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-Related Disease Research, Central South University, Changsha, China
| | - Shuang Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-Related Disease Research, Central South University, Changsha, China
| | - You-Shuo Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Aging and Age-Related Disease Research, Central South University, Changsha, China
| |
Collapse
|
11
|
Accelerated DNA methylation age and medication use among African Americans. Aging (Albany NY) 2021; 13:14604-14629. [PMID: 34083497 PMCID: PMC8221348 DOI: 10.18632/aging.203115] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 05/14/2021] [Indexed: 12/16/2022]
Abstract
DNA methylation age acceleration, the discrepancy between epigenetic age and chronological age, is associated with mortality and chronic diseases, including diabetes, hypertension, and hyperlipidemia. In this study, we investigate whether medications commonly used to treat these diseases in 15 drug categories are associated with four epigenetic age acceleration measures: HorvathAge acceleration (HorvathAA), HannumAge acceleration (HannumAA), PhenoAge acceleration, and GrimAge acceleration (GrimAA) using cross-sectional (Phase 1, N=1,100) and longitudinal (Phases 1 and 2, N=266) data from African Americans in the Genetic Epidemiology Network of Arteriopathy (GENOA) study. In cross-sectional analyses, the use of calcium channel blockers was associated with 1.27 years lower HannumAA after adjusting for covariates including hypertension (p=0.001). Longitudinal analyses showed that, compared to those who never used antihypertensives, those who started to take antihypertensives after Phase 1 had a 0.97-year decrease in GrimAA (p=0.007). In addition, compared to those who never used NSAID analgesics, those who started to take them after Phase 1 had a 2.61-year increase in HorvathAA (p=0.0005). Our study demonstrates that three commonly used medications are associated with DNAm age acceleration in African Americans and sheds light on the potential epigenetic effects of pharmaceuticals on aging at the cellular level.
Collapse
|
12
|
Hong C, Yang S, Wang Q, Zhang S, Wu W, Chen J, Zhong D, Li M, Li L, Li J, Yu H, Chen H, Zeng Q, Zhang C. Epigenetic Age Acceleration of Stomach Adenocarcinoma Associated With Tumor Stemness Features, Immunoactivation, and Favorable Prognosis. Front Genet 2021; 12:563051. [PMID: 33815458 PMCID: PMC8012546 DOI: 10.3389/fgene.2021.563051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 02/22/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Abnormal DNA methylation (DNAm) age has been assumed to be an indicator for canceration and all-cause mortality. However, associations between DNAm age and molecular features of stomach adenocarcinoma (STAD), and its prognosis have not been systematically studied. Method: We calculated the DNAm age of 591 STAD samples and 115 normal stomach samples from The Cancer Genome Atlas (TCGA) and gene expression omnibus (GEO) database using the Horvath’s clock model. Meanwhile, we utilized survival analysis to evaluate the prognostic value of DNAm age and epigenetic age acceleration shift. In addition, we performed weighted gene co-expression network analysis (WGCNA) to identify DNAm age-associated gene modules and pathways. Finally, the association between DNAm age and molecular features was performed by correlation analysis. Results: DNA methylation age was significantly correlated with chronological age in normal gastric tissues (r = 0.85, p < 0.0001), but it was not associated with chronological age in STAD samples (r = 0.060, p = 0.2369). Compared with tumor adjacent normal tissue, the DNAm age of STAD tissues was significantly decreased. Meanwhile, chronological age in STAD samples was higher than its DNAm age. Both DNAm age and epigenetic acceleration shift were associated with the prognosis of STAD patients. By using correlation analysis, we also found that DNAm age was associated with immunoactivation and stemness in STAD samples. Conclusion: In summary, epigenetic age acceleration of STAD was associated with tumor stemness, immunoactivation, and favorable prognosis.
Collapse
Affiliation(s)
- Chunhong Hong
- Center of Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Shaohua Yang
- Center of Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Qiaojin Wang
- Department of Surgical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shiqiang Zhang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Wenhui Wu
- Center of Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jinyao Chen
- Center of Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Danhui Zhong
- Department of Physiotherapy, The University of Hongkong-Shenzhen Hospital, Shenzhen, China
| | - Mingzhe Li
- Center of Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Liang Li
- Center of Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jianfeng Li
- Center of Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hong Yu
- Center of Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hong Chen
- Center of Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Qianlin Zeng
- Center of Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Changhua Zhang
- Center of Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
13
|
Prada D, Belsky D, Baccarelli A. Is your environment making you older? Molecular biomarkers and new approaches to investigate the influences of environmental chemicals through aging. LA MEDICINA DEL LAVORO 2021; 112:8-14. [PMID: 33635291 PMCID: PMC8023055 DOI: 10.23749/mdl.v112i1.10826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/12/2020] [Indexed: 12/14/2022]
Abstract
Aging is characterized by a gradual and progressive decline in system integrity that occurs with advancing chronological age. Although it is a physiological process, aging is associated with a myriad of age-related diseases (ARDs), including frailty, sarcopenia, chronic obstructive pulmonary disease, cardiovascular disease, cancer, and neurodegenerative diseases. While not exclusively ARDs, many of these diseases lead to death, a lesser quality of life, and increased healthcare costs for individuals and systems. ARDs share several underlying molecular mechanisms, such as cellular damage, inflammation, DNA methylation changes, stem cells exhaustion, and DNA mutations, which have been outlined as hallmarks of aging. Evidence suggests that environmental exposures, including but not limited to metals, air pollution, endocrine-disrupting chemicals, and noise, may accelerate biological aging. Over the past few years, aging research has identified new molecular biomarkers of the aging process. When applied to investigate environmental influences, these biomarkers can help identify individuals who are particularly susceptible to the influences of environmental exposures on aging processes and therefore guide in implementing possible preventive measures.
Collapse
Affiliation(s)
- Diddier Prada
- Mailman School of Public Health, Columbia University, USA; Instituto Nacional de Cancerología, Mexico.
| | - Daniel Belsky
- Columbia University Mailman School of Public Health .
| | | |
Collapse
|
14
|
Deng Q, Su B, Ji X, Fang Q, Zhou S, Zhou C. Predictive value of unmethylated RASSF1A on disease progression in non-small cell lung cancer patients receiving pemetrexed-based chemotherapy. Cancer Biomark 2020; 27:313-323. [PMID: 31839603 DOI: 10.3233/cbm-190258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND OBJECTIVE Chemotherapy remains the basis of the treatment of lung cancer, and screening biomarkers with predictive value for chemotherapy is of great interest. The present study focused on status of genes methylation in NSCLC patients receiving pemetrexed- or gemcitabine-based chemotherapy. PATIENTS AND METHODS Promoter methylation of Ras association domain family (RASSF1A) and short stature homeobox 2 (SHOX2) was examined in bronchoalveolar lavage (BAL) from 117 NSCLC patients treated with chemotherapy. Multivariate analysis was used to identify the predictive value of gene methylation. Progression-free survival (PFS) rather than overall survival (OS) was used as the clinical outcome to minimize the impact of chemotherapy on gene methylation. RESULTS The methylation of RASSF1A and SHOX2 was significantly associated with shorter PFS (RASSF1A: HR = 2.355, 95% CI: 1.533-3.617, P< 0.0001; SHOX2: HR = 2.123, 95% CI: 1.392-3.236, P= 0.0004). After adjusting for confounding factors, RASSF1A methylation was still a predictive factor for PFS (HR = 1.765, 95% CI: 1.064-2.928, P= 0.0278). In the pemetrexed group, unmethylated RASSF1A could be used to predict longer PFS (P= 0.0001), and no predictive value was found in the gemcitabine group. CONCLUSION Unmethylated RASSF1A is a favorable prognostic indicator for patients receiving pemetrexed doublets. Because of the promoting effect of most chemotherapeutic drugs on gene methylation, unmethylated RASSF1A is not suitable as a predictor for gemcitabine doublets.
Collapse
Affiliation(s)
- Qinfang Deng
- Medical College of Soochow University, Soochow, China.,Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.,Medical College of Soochow University, Soochow, China
| | - Bo Su
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.,Medical College of Soochow University, Soochow, China
| | - Xianxiu Ji
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiyu Fang
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Songwen Zhou
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Caicun Zhou
- Medical College of Soochow University, Soochow, China.,Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Accelerated epigenetic aging as a risk factor for chronic obstructive pulmonary disease and decreased lung function in two prospective cohort studies. Aging (Albany NY) 2020; 12:16539-16554. [PMID: 32747609 PMCID: PMC7485704 DOI: 10.18632/aging.103784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/14/2020] [Indexed: 12/14/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a frequent diagnosis in older individuals and contributor to global morbidity and mortality. Given the link between lung disease and aging, we need to understand how molecular indicators of aging relate to lung function and disease. Using data from the population-based KORA (Cooperative Health Research in the Region of Augsburg) surveys, we associated baseline epigenetic (DNA methylation) age acceleration with incident COPD and lung function. Models were adjusted for age, sex, smoking, height, weight, and baseline lung disease as appropriate. Associations were replicated in the Normative Aging Study. Of 770 KORA participants, 131 developed incident COPD over 7 years. Baseline accelerated epigenetic aging was significantly associated with incident COPD. The change in age acceleration (follow-up - baseline) was more strongly associated with COPD than baseline aging alone. The association between the change in age acceleration between baseline and follow-up and incident COPD replicated in the Normative Aging Study. Associations with spirometric lung function parameters were weaker than those with COPD, but a meta-analysis of both cohorts provide suggestive evidence of associations. Accelerated epigenetic aging, both baseline measures and changes over time, may be a risk factor for COPD and reduced lung function.
Collapse
|
16
|
Belsky DW, Caspi A, Arseneault L, Baccarelli A, Corcoran DL, Gao X, Hannon E, Harrington HL, Rasmussen LJH, Houts R, Huffman K, Kraus WE, Kwon D, Mill J, Pieper CF, Prinz JA, Poulton R, Schwartz J, Sugden K, Vokonas P, Williams BS, Moffitt TE. Quantification of the pace of biological aging in humans through a blood test, the DunedinPoAm DNA methylation algorithm. eLife 2020; 9:e54870. [PMID: 32367804 PMCID: PMC7282814 DOI: 10.7554/elife.54870] [Citation(s) in RCA: 293] [Impact Index Per Article: 58.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022] Open
Abstract
Biological aging is the gradual, progressive decline in system integrity that occurs with advancing chronological age, causing morbidity and disability. Measurements of the pace of aging are needed as surrogate endpoints in trials of therapies designed to prevent disease by slowing biological aging. We report a blood-DNA-methylation measure that is sensitive to variation in pace of biological aging among individuals born the same year. We first modeled change-over-time in 18 biomarkers tracking organ-system integrity across 12 years of follow-up in n = 954 members of the Dunedin Study born in 1972-1973. Rates of change in each biomarker over ages 26-38 years were composited to form a measure of aging-related decline, termed Pace-of-Aging. Elastic-net regression was used to develop a DNA-methylation predictor of Pace-of-Aging, called DunedinPoAm for Dunedin(P)ace(o)f(A)ging(m)ethylation. Validation analysis in cohort studies and the CALERIE trial provide proof-of-principle for DunedinPoAm as a single-time-point measure of a person's pace of biological aging.
Collapse
Affiliation(s)
- Daniel W Belsky
- Department of Epidemiology, Columbia University Mailman School of Public HealthNew YorkUnited States
- Butler Columbia Aging Center, Columbia University Mailman School of Public HealthNew YorkUnited States
| | - Avshalom Caspi
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, King’s College LondonLondonUnited Kingdom
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
- Department of Psychiatry and Behavioral Sciences, Duke University School of MedicineDurhamUnited States
- Center for Genomic and Computational Biology, Duke UniversityDurhamUnited States
| | - Louise Arseneault
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, King’s College LondonLondonUnited Kingdom
| | - Andrea Baccarelli
- Laboratory of Precision Environmental Health, Mailman School of Public Health, Columbia UniversityNew YorkUnited States
| | - David L Corcoran
- Center for Genomic and Computational Biology, Duke UniversityDurhamUnited States
| | - Xu Gao
- Laboratory of Precision Environmental Health, Mailman School of Public Health, Columbia UniversityNew YorkUnited States
| | - Eiliss Hannon
- University of Exeter Medical School, College of Medicine and HealthExeterUnited Kingdom
| | - Hona Lee Harrington
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Line JH Rasmussen
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
- Clinical Research Centre, Copenhagen University Hospital Amager and HvidovreHvidovreDenmark
| | - Renate Houts
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Kim Huffman
- Duke Molecular Physiology Institute, Duke UniversityDurhamUnited States
- Duke University Center for the Study of Aging, Duke UniversityDurhamUnited States
| | - William E Kraus
- Duke Molecular Physiology Institute, Duke UniversityDurhamUnited States
- Duke University Center for the Study of Aging, Duke UniversityDurhamUnited States
| | - Dayoon Kwon
- Butler Columbia Aging Center, Columbia University Mailman School of Public HealthNew YorkUnited States
| | - Jonathan Mill
- University of Exeter Medical School, College of Medicine and HealthExeterUnited Kingdom
| | - Carl F Pieper
- Duke University Center for the Study of Aging, Duke UniversityDurhamUnited States
- Department of Biostatistics, Duke University School of MedicineDurhamUnited States
| | - Joseph A Prinz
- Center for Genomic and Computational Biology, Duke UniversityDurhamUnited States
| | - Richie Poulton
- Department of Psychology and Dunedin Multidisciplinary Health and Development Research Unit, University of OtagoOtagoNew Zealand
| | - Joel Schwartz
- Department of Environmental Health Sciences, Harvard TH Chan School of Public HealthBostonUnited States
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Pantel Vokonas
- Veterans Affairs Normative Aging Study, Veterans Affairs Boston Healthcare System, Department of Medicine, Boston University School of MedicineBostonUnited States
| | - Benjamin S Williams
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
| | - Terrie E Moffitt
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, King’s College LondonLondonUnited Kingdom
- Department of Psychology and Neuroscience, Duke UniversityDurhamUnited States
- Department of Psychiatry and Behavioral Sciences, Duke University School of MedicineDurhamUnited States
- Center for Genomic and Computational Biology, Duke UniversityDurhamUnited States
| |
Collapse
|
17
|
Hu J, Zhu H, Xu G, Chen Z, Li L, Wang S, Deng H, Bao X, Shen Z. Significant association between DHFR promoter methylation and ischemic stroke in a Chinese hypertensive population. J Clin Lab Anal 2020; 34:e23322. [PMID: 32319147 PMCID: PMC7439332 DOI: 10.1002/jcla.23322] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 03/09/2020] [Accepted: 03/11/2020] [Indexed: 12/22/2022] Open
Abstract
Objective DHFR encodes dihydrofolate reductase, a major enzyme in the metabolism of folate, and is a candidate gene for ischemic stroke (IS). Therefore, we aimed to investigate the association between DHFR promoter methylation and IS in a Chinese population with primary hypertension. Methods Quantitative methylation‐specific PCR was used to measure the level of DHFR promoter methylation. A multivariate logistic regression model was used to investigate the association between DHFR promoter methylation and IS. Receiver operating characteristic (ROC) curve analysis was used to evaluate the diagnostic value of DHFR promoter methylation for IS. Results The level of methylation of the DHFR promoter in the IS group was significantly lower than that in the hypertensive group (median [interquartile range]: 9.11 [2.81‐16.20] vs 24.94 [7.16‐56.45], P < .001). DHFR promoter methylation and homocysteine (Hcy) levels were both related to IS, with an ORs (95% CI) of 0.976 (0.967‐0.984) and 1.057 (1.027‐1.108), respectively. The areas under the curve for the diagnosis of DHFR promoter hypomethylation in IS were 0.603 (95% CI, 0.527‐0.678) in men and 0.754 (95% CI, 0.693‐0.815) in women. A dual‐luciferase reporter assay revealed that the target sequence in the DHFR promoter upregulated gene expression. Conclusion There is a significant association between methylation of the DHFR promoter and IS in this Chinese hypertensive population. Hypomethylation of the DHFR promoter may serve as a novel marker for the diagnosis of IS in women.
Collapse
Affiliation(s)
- Jingcen Hu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Hong Zhu
- Gynecology & Obstetrics Department, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Guodong Xu
- Medical Record Statistics Room, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Zhu Chen
- HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Lian Li
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Shuyu Wang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Hongxia Deng
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Xiaoming Bao
- HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China.,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Zhisen Shen
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| |
Collapse
|
18
|
Gao X, Colicino E, Shen J, Just AC, Nwanaji-Enwerem JC, Wang C, Coull B, Lin X, Vokonas P, Zheng Y, Hou L, Schwartz J, Baccarelli AA. Comparative validation of an epigenetic mortality risk score with three aging biomarkers for predicting mortality risks among older adult males. Int J Epidemiol 2019; 48:1958-1971. [PMID: 31038702 PMCID: PMC6929530 DOI: 10.1093/ije/dyz082] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND A 'mortality risk score' (MS) based on ten prominent mortality-related cytosine-phosphate-guanine (CpG) sites was previously associated with all-cause mortality, but has not been verified externally. We aimed to validate the association of MS with mortality and to compare MS with three aging biomarkers: telomere length (TL), DNA methylation age (DNAmAge) and phenotypic age (DNAmPhenoAge) to explore whether MS can serve as a reliable measure of biological aging and mortality. METHODS Among 534 males aged 55-85 years from the US Normative Aging Study, the MS, DNAmAge and DNAmPhenoAge were derived from blood DNA methylation profiles from the Illumina HumanMethylation450 BeadChip, and TL was measured by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS A total of 147 participants died during a median follow-up of 9.4 years. The MS showed strong associations with all-cause, cardiovascular disease (CVD) and cancer mortality. After controlling for all potential covariates, participants with high MS (>5 CpG sites with aberrant methylation) had almost 4-fold all-cause mortality (hazard ratio: 3.84, 95% confidence interval: 1.92-7.67) compared with participants with a low MS (0-1 CpG site with aberrant methylation). Similar patterns were observed with respect to CVD and cancer mortality. MS was associated with TL and DNAmPhenoAge acceleration but not with DNAmAge acceleration. Although the MS and DNAmPhenoAge acceleration were independently associated with all-cause mortality, the former exhibited a higher predictive accuracy of mortality than the latter. CONCLUSIONS MS has the potential to be a prominent predictor of mortality that could enhance survival prediction in clinical settings.
Collapse
Affiliation(s)
- Xu Gao
- Laboratory of Precision Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Elena Colicino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jincheng Shen
- Department of Population Health Sciences, University of Utah, School of Medicine, Salt Lake City, UT, USA
| | - Allan C Just
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Cuicui Wang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Brent Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Pantel Vokonas
- Veterans Affairs Normative Aging Study, Veterans Affairs Boston Healthcare System, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Yinan Zheng
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lifang Hou
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joel Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andrea A Baccarelli
- Laboratory of Precision Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
19
|
Gao X, Colicino E, Shen J, Kioumourtzoglou MA, Just AC, Nwanaji-Enwerem JC, Coull B, Lin X, Vokonas P, Zheng Y, Hou L, Schwartz J, Baccarelli AA. Impacts of air pollution, temperature, and relative humidity on leukocyte distribution: An epigenetic perspective. ENVIRONMENT INTERNATIONAL 2019; 126:395-405. [PMID: 30826618 PMCID: PMC6441628 DOI: 10.1016/j.envint.2019.02.053] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/15/2019] [Accepted: 02/21/2019] [Indexed: 05/20/2023]
Abstract
BACKGROUND Exploring the associations of air pollution and weather variables with blood leukocyte distribution is critical to understand the impacts of environmental exposures on the human immune system. OBJECTIVES As previous analyses have been mainly based on data from cell counters, which might not be feasible in epidemiologic studies including large populations of long-stored blood samples, we aimed to expand the understanding of this topic by employing the leukocyte distribution estimated by DNA methylation profiles. METHODS We measured DNA methylation profiles in blood samples using Illumina HumanMethylation450 BeadChip from 1519 visits of 774 Caucasian males participating in the Normative Aging Study. Leukocyte distribution was estimated using Houseman's and Horvath's algorithms. Data on air pollution exposure, temperature, and relative humidity within 28 days before each blood draw was obtained. RESULTS After fully adjusting for potential covariates, PM2.5, black carbon, particle number, carbon monoxide, nitrogen dioxide, sulfur dioxide, temperature, and relative humidity were associated with the proportions of at least one subtype of leukocytes. Particularly, an interquartile range-higher 28-day average exposure of PM2.5 was associated with 0.147-, 0.054- and 0.101-unit lower proportions (z-scored) of plasma cells, naïve CD8+ T cells, and natural killers, respectively, and 0.059- and 0.161-unit higher proportions (z-scored) of naïve CD4+ T cells and CD8+ T cells, respectively. CONCLUSIONS Our study suggests that short-term air pollution exposure, temperature, and relative humidity are associated with leukocyte distribution. Our study further provides a successful attempt to use epigenetic patterns to assess the influences of environmental exposures on human immune profiles.
Collapse
Affiliation(s)
- Xu Gao
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA.
| | - Elena Colicino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jincheng Shen
- Department of Population Health Sciences, University of Utah, School of Medicine, Salt Lake City, UT, USA
| | | | - Allan C Just
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Brent Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Pantel Vokonas
- Veterans Affairs Normative Aging Study, Veterans Affairs Boston Healthcare System, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Yinan Zheng
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Lifang Hou
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - 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
| |
Collapse
|