1
|
Shealy EP, Schwartz TS, Cox RM, Reedy AM, Parrott BB. DNA methylation-based age prediction and sex-specific epigenetic aging in a lizard with female-biased longevity. SCIENCE ADVANCES 2025; 11:eadq3589. [PMID: 39888991 PMCID: PMC11784858 DOI: 10.1126/sciadv.adq3589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 01/02/2025] [Indexed: 02/02/2025]
Abstract
Sex differences in life span are widespread across animal taxa, but their causes remain unresolved. Alterations to the epigenome are hypothesized to contribute to vertebrate aging, and DNA methylation-based aging clocks allow for quantitative estimation of biological aging trajectories. Here, we investigate the influence of age, sex, and their interaction on genome-wide DNA methylation patterns in the brown anole (Anolis sagrei), a lizard with pronounced female-biased survival and longevity. We develop a series of age predictor models and find that, contrary to our predictions, rates of epigenetic aging were not slower in female lizards. However, methylation states at loci acquiring age-associated changes appear to be more "youthful" in young females, suggesting that female DNA methylomes are preemptively fortified in early life in opposition to the direction of age-related drift. Collectively, our findings provide insights into epigenetic aging in reptiles and suggest that early-life epigenetic profiles may be more informative than rates of change for predicting sex biases in longevity.
Collapse
Affiliation(s)
- Ethan P. Shealy
- Savannah River Ecology Laboratory, University of Georgia, Aiken, SC 29802, USA
- Eugene P. Odum School of Ecology, University of Georgia, Athens, GA 30602, USA
- Interdisciplinary Toxicology Program, University of Georgia, Athens, GA 30602, USA
| | - Tonia S. Schwartz
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Robert M. Cox
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Aaron M. Reedy
- DataClassroom, 1022 Cottonwood Rd, Charlottesville, VA 22901, USA
| | - Benjamin B. Parrott
- Savannah River Ecology Laboratory, University of Georgia, Aiken, SC 29802, USA
- Eugene P. Odum School of Ecology, University of Georgia, Athens, GA 30602, USA
- Interdisciplinary Toxicology Program, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
2
|
Olova NN. Epigenetic rejuvenation: a journey backwards towards an epigenomic ground state. Epigenomics 2025; 17:1-3. [PMID: 39584805 DOI: 10.1080/17501911.2024.2432851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024] Open
Affiliation(s)
- Nelly N Olova
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
- Institute of Biodiversity, Animal Health and Comparative Medicine, School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| |
Collapse
|
3
|
Kriukov D, Kuzmina E, Efimov E, Dylov DV, Khrameeva EE. Epistemic uncertainty challenges aging clock reliability in predicting rejuvenation effects. Aging Cell 2024; 23:e14283. [PMID: 39072888 PMCID: PMC11561706 DOI: 10.1111/acel.14283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
Epigenetic aging clocks have been widely used to validate rejuvenation effects during cellular reprogramming. However, these predictions are unverifiable because the true biological age of reprogrammed cells remains unknown. We present an analytical framework to consider rejuvenation predictions from the uncertainty perspective. Our analysis reveals that the DNA methylation profiles across reprogramming are poorly represented in the aging data used to train clock models, thus introducing high epistemic uncertainty in age estimations. Moreover, predictions of different published clocks are inconsistent, with some even suggesting zero or negative rejuvenation. While not questioning the possibility of age reversal, we show that the high clock uncertainty challenges the reliability of rejuvenation effects observed during in vitro reprogramming before pluripotency and throughout embryogenesis. Conversely, our method reveals a significant age increase after in vivo reprogramming. We recommend including uncertainty estimation in future aging clock models to avoid the risk of misinterpreting the results of biological age prediction.
Collapse
Affiliation(s)
- Dmitrii Kriukov
- Skolkovo Institute of Science and TechnologyMoscowRussia
- Artificial Intelligence Research InstituteMoscowRussia
| | - Ekaterina Kuzmina
- Skolkovo Institute of Science and TechnologyMoscowRussia
- Artificial Intelligence Research InstituteMoscowRussia
| | - Evgeniy Efimov
- Skolkovo Institute of Science and TechnologyMoscowRussia
| | - Dmitry V. Dylov
- Skolkovo Institute of Science and TechnologyMoscowRussia
- Artificial Intelligence Research InstituteMoscowRussia
| | | |
Collapse
|
4
|
Zakar-Polyák E, Csordas A, Pálovics R, Kerepesi C. Profiling the transcriptomic age of single-cells in humans. Commun Biol 2024; 7:1397. [PMID: 39462118 PMCID: PMC11513945 DOI: 10.1038/s42003-024-07094-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Although aging clocks predicting the age of individual organisms have been extensively studied, the age of individual cells remained largely unexplored. Most recently single-cell omics clocks were developed for the mouse, however, extensive profiling the age of human cells is still lacking. To fill this gap, here we use available scRNA-seq data of 1,058,909 blood cells of 508 healthy, human donors (between 19 and 75 years), for developing single-cell transcriptomic clocks and predicting the age of human blood cells. By the application of the proposed cell-type-specific single-cell clocks, our main observations are that (i) transcriptomic age is associated with cellular senescence; (ii) the transcriptomic age of classical monocytes as well as naive B and T cells is decreased in moderate COVID-19 followed by an increase for some cell types in severe COVID-19; and (iii) the human embryo cells transcriptomically rejuvenated at the morulae and blastocyst stages. In summary, here we demonstrate that single-cell transcriptomic clocks are useful tools to investigate aging and rejuvenation at the single-cell level.
Collapse
Affiliation(s)
- Enikő Zakar-Polyák
- Institute for Computer Science and Control (SZTAKI), Hungarian Research Network (HUN-REN), Budapest, Hungary.
- Doctoral School of Informatics, Eötvös Loránd University, Budapest, Hungary.
| | - Attila Csordas
- AgeCurve Limited, Cambridge, UK
- Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
| | - Róbert Pálovics
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Csaba Kerepesi
- Institute for Computer Science and Control (SZTAKI), Hungarian Research Network (HUN-REN), Budapest, Hungary.
| |
Collapse
|
5
|
Meyer DH, Schumacher B. Aging clocks based on accumulating stochastic variation. NATURE AGING 2024; 4:871-885. [PMID: 38724736 PMCID: PMC11186771 DOI: 10.1038/s43587-024-00619-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 03/28/2024] [Indexed: 05/15/2024]
Abstract
Aging clocks have provided one of the most important recent breakthroughs in the biology of aging, and may provide indicators for the effectiveness of interventions in the aging process and preventive treatments for age-related diseases. The reproducibility of accurate aging clocks has reinvigorated the debate on whether a programmed process underlies aging. Here we show that accumulating stochastic variation in purely simulated data is sufficient to build aging clocks, and that first-generation and second-generation aging clocks are compatible with the accumulation of stochastic variation in DNA methylation or transcriptomic data. We find that accumulating stochastic variation is sufficient to predict chronological and biological age, indicated by significant prediction differences in smoking, calorie restriction, heterochronic parabiosis and partial reprogramming. Although our simulations may not explicitly rule out a programmed aging process, our results suggest that stochastically accumulating changes in any set of data that have a ground state at age zero are sufficient for generating aging clocks.
Collapse
Affiliation(s)
- David H Meyer
- Institute for Genome Stability in Aging and Disease, University Hospital and University of Cologne, Cologne, Germany.
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| | - Björn Schumacher
- Institute for Genome Stability in Aging and Disease, University Hospital and University of Cologne, Cologne, Germany.
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
6
|
Park SH, Lee DH, Lee DH, Jung CH. Scientific evidence of foods that improve the lifespan and healthspan of different organisms. Nutr Res Rev 2024; 37:169-178. [PMID: 37469212 DOI: 10.1017/s0954422423000136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Age is a risk factor for numerous diseases. Although the development of modern medicine has greatly extended the human lifespan, the duration of relatively healthy old age, or 'healthspan', has not increased. Targeting the detrimental processes that can occur before the onset of age-related diseases can greatly improve health and lifespan. Healthspan is significantly affected by what, when and how much one eats. Dietary restriction, including calorie restriction, fasting or fasting-mimicking diets, to extend both lifespan and healthspan has recently attracted much attention. However, direct scientific evidence that consuming specific foods extends the lifespan and healthspan seems lacking. Here, we synthesized the results of recent studies on the lifespan and healthspan extension properties of foods and their phytochemicals in various organisms to confirm how far the scientific research on the effect of food on the lifespan has reached.
Collapse
Affiliation(s)
- So-Hyun Park
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, South Korea
| | - Da-Hye Lee
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Dae-Hee Lee
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung, Gangwon-do, South Korea
| | - Chang Hwa Jung
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, South Korea
- Department of Food Biotechnology, University of Science and Technology, Wanju-gun, Jeollabuk-do, South Korea
| |
Collapse
|
7
|
Niimi P, Gould V, Thrush-Evensen K, Levine ME. The Latent Aging of Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596284. [PMID: 38854054 PMCID: PMC11160607 DOI: 10.1101/2024.05.28.596284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
As epigenetic clocks have evolved from powerful estimators of chronological aging to predictors of mortality and disease risk, it begs the question of what role DNA methylation plays in the aging process. We hypothesize that while it has the potential to serve as an informative biomarker, DNA methylation could also be a key to understanding the biology entangled between aging, (de)differentiation, and epigenetic reprogramming. Here we use an unsupervised approach to analyze time associated DNA methylation from both in vivo and in vitro samples to measure an underlying signal that ties these phenomena together. We identify a methylation pattern shared across all three, as well as a signal that tracks aging in tissues but appears refractory to reprogramming, suggesting that aging and reprogramming may not be fully mirrored processes.
Collapse
Affiliation(s)
- Peter Niimi
- Program in Experimental Pathology, Yale University, New Haven, CT, USA
- Altos Labs, San Diego Institute of Science, San Diego, CA, USA
| | - Victoria Gould
- Altos Labs, San Diego Institute of Science, San Diego, CA, USA
| | | | - Morgan E Levine
- Altos Labs, San Diego Institute of Science, San Diego, CA, USA
| |
Collapse
|
8
|
Loseva PA, Gladyshev VN. The beginning of becoming a human. Aging (Albany NY) 2024; 16:8378-8395. [PMID: 38713165 PMCID: PMC11131989 DOI: 10.18632/aging.205824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/27/2024] [Indexed: 05/08/2024]
Abstract
According to birth certificates, the life of a child begins once their body comes out of the mother's womb. But when does their organismal life begin? Science holds a palette of answers-depending on how one defines a human life. In 1984, a commission on the regulatory framework for human embryo experimentation opted not to answer this question, instead setting a boundary, 14 days post-fertilization, beyond which any experiments were forbidden. Recently, as the reproductive technologies developed and the demand for experimentation grew stronger, this boundary may be set aside leaving the ultimate decision to local oversight committees. While science has not come closer to setting a zero point for human life, there has been significant progress in our understanding of early mammalian embryogenesis. It has become clear that the 14-day stage does in fact possess features, which make it a foundational time point for a developing human. Importantly, this stage defines the separation of soma from the germline and marks the boundary between rejuvenation and aging. We explore how different levels of life organization emerge during human development and suggest a new meaning for the 14-day stage in organismal life that is grounded in recent mechanistic advances and insights from aging studies.
Collapse
Affiliation(s)
- Polina A. Loseva
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
9
|
Mitchell W, Goeminne LJE, Tyshkovskiy A, Zhang S, Chen JY, Paulo JA, Pierce KA, Choy AH, Clish CB, Gygi SP, Gladyshev VN. Multi-omics characterization of partial chemical reprogramming reveals evidence of cell rejuvenation. eLife 2024; 12:RP90579. [PMID: 38517750 PMCID: PMC10959535 DOI: 10.7554/elife.90579] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024] Open
Abstract
Partial reprogramming by cyclic short-term expression of Yamanaka factors holds promise for shifting cells to younger states and consequently delaying the onset of many diseases of aging. However, the delivery of transgenes and potential risk of teratoma formation present challenges for in vivo applications. Recent advances include the use of cocktails of compounds to reprogram somatic cells, but the characteristics and mechanisms of partial cellular reprogramming by chemicals remain unclear. Here, we report a multi-omics characterization of partial chemical reprogramming in fibroblasts from young and aged mice. We measured the effects of partial chemical reprogramming on the epigenome, transcriptome, proteome, phosphoproteome, and metabolome. At the transcriptome, proteome, and phosphoproteome levels, we saw widescale changes induced by this treatment, with the most notable signature being an upregulation of mitochondrial oxidative phosphorylation. Furthermore, at the metabolome level, we observed a reduction in the accumulation of aging-related metabolites. Using both transcriptomic and epigenetic clock-based analyses, we show that partial chemical reprogramming reduces the biological age of mouse fibroblasts. We demonstrate that these changes have functional impacts, as evidenced by changes in cellular respiration and mitochondrial membrane potential. Taken together, these results illuminate the potential for chemical reprogramming reagents to rejuvenate aged biological systems and warrant further investigation into adapting these approaches for in vivo age reversal.
Collapse
Affiliation(s)
- Wayne Mitchell
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Ludger JE Goeminne
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Sirui Zhang
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Julie Y Chen
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
| | - Kerry A Pierce
- Broad Institute of MIT and HarvardCambridgeUnited States
| | | | - Clary B Clish
- Broad Institute of MIT and HarvardCambridgeUnited States
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
10
|
Raffington L. Utilizing epigenetics to study the shared nature of development and biological aging across the lifespan. NPJ SCIENCE OF LEARNING 2024; 9:24. [PMID: 38509146 PMCID: PMC10954727 DOI: 10.1038/s41539-024-00239-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 03/13/2024] [Indexed: 03/22/2024]
Abstract
Recently, biological aging has been quantified in DNA-methylation samples of older adults and applied as so-called "methylation profile scores" (MPSs) in separate target samples, including samples of children. This nascent research indicates that (1) biological aging can be quantified early in the life course, decades before the onset of aging-related disease, (2) is affected by common environmental predictors of childhood development, and (3) shows overlap with "developmental processes" (e.g., puberty). Because the MPSs were computed using algorithms developed in adults, these studies indicate a molecular link between childhood environments, development, and adult biological aging. Yet, if MPSs can be used to connect development and aging, previous research has only traveled one way, deriving MPSs developed in adults and applying them to samples of children. Researchers have not yet quantified epigenetic measures that reflect the pace of child development, and tested whether resulting MPSs are associated with physical and psychological aging. In this perspective I posit that combining measures of biological aging with new quantifications of child development has the power to address fundamental questions about life span: How are development and experience in childhood related to biological aging in adulthood? And what is aging?
Collapse
Affiliation(s)
- Laurel Raffington
- Max Planck Research Group Biosocial-Biology, Social Disparities, and Development, Max Planck Institute for Human Development, Lentzeallee 94, 14195, Berlin, Germany.
| |
Collapse
|
11
|
Riddle NC, Biga PR, Bronikowski AM, Walters JR, Wilkinson GS. Comparative analysis of animal lifespan. GeroScience 2024; 46:171-181. [PMID: 37889438 PMCID: PMC10828364 DOI: 10.1007/s11357-023-00984-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 10/15/2023] [Indexed: 10/28/2023] Open
Abstract
Comparative studies of aging are a promising approach to identifying general properties of and processes leading to aging. While to date, many comparative studies of aging in animals have focused on relatively narrow species groups, methodological innovations now allow for studies that include evolutionary distant species. However, comparative studies of aging across a wide range of species that have distinct life histories introduce additional challenges in experimental design. Here, we discuss these challenges, highlight the most pressing problems that need to be solved, and provide suggestions based on current approaches to successfully carry out comparative aging studies across the animal kingdom.
Collapse
Affiliation(s)
- Nicole C Riddle
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Peggy R Biga
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anne M Bronikowski
- Department of Integrative Biology, Kellogg Biological Station, Michigan State University, Hickory Corners, MI, USA
| | - James R Walters
- Department of Ecology and Evolutionary Biology, The University of Kansas, Lawrence, KS, USA
| | | |
Collapse
|
12
|
Etzel L, Garrett-Petters P, Shalev I. Early origins of health and disease risk: The case for investigating adverse exposures and biological aging in utero, across childhood, and into adolescence. CHILD DEVELOPMENT PERSPECTIVES 2023; 17:149-156. [PMID: 38706692 PMCID: PMC11068077 DOI: 10.1111/cdep.12488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
In this article, we suggest that aging and development are two sides of the same coin, and that developing a comprehensive understanding of health and disease risk requires examining age-related processes occurring throughout the earliest years of life. Compared to other periods in life, during this early period of acute vulnerability, when children's biological and regulatory systems are developing, biological aging occurs most rapidly. We review theory and empirical research suggesting that processes of development and aging are intricately linked, and that early adversity may program biological parameters for accelerated aging and disease risk early in life, even though clinical signs of age-related disease onset may not be evident until many years later. Following from this, we make the case for widespread incorporation of biological aging constructs into child development research.
Collapse
Affiliation(s)
- Laura Etzel
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, USA
| | | | - Idan Shalev
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
13
|
Mitchell W, Goeminne LJ, Tyshkovskiy A, Zhang S, Chen JY, Paulo JA, Pierce KA, Choy AH, Clish CB, Gygi SP, Gladyshev VN. Multi-omics characterization of partial chemical reprogramming reveals evidence of cell rejuvenation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.30.546730. [PMID: 37425825 PMCID: PMC10327104 DOI: 10.1101/2023.06.30.546730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Partial reprogramming by cyclic short-term expression of Yamanaka factors holds promise for shifting cells to younger states and consequently delaying the onset of many diseases of aging. However, the delivery of transgenes and potential risk of teratoma formation present challenges for in vivo applications. Recent advances include the use of cocktails of compounds to reprogram somatic cells, but the characteristics and mechanisms of partial cellular reprogramming by chemicals remain unclear. Here, we report a multi-omics characterization of partial chemical reprogramming in fibroblasts from young and aged mice. We measured the effects of partial chemical reprogramming on the epigenome, transcriptome, proteome, phosphoproteome, and metabolome. At the transcriptome, proteome, and phosphoproteome levels, we saw widescale changes induced by this treatment, with the most notable signature being an upregulation of mitochondrial oxidative phosphorylation. Furthermore, at the metabolome level, we observed a reduction in the accumulation of aging-related metabolites. Using both transcriptomic and epigenetic clock-based analyses, we show that partial chemical reprogramming reduces the biological age of mouse fibroblasts. We demonstrate that these changes have functional impacts, as evidenced by changes in cellular respiration and mitochondrial membrane potential. Taken together, these results illuminate the potential for chemical reprogramming reagents to rejuvenate aged biological systems and warrant further investigation into adapting these approaches for in vivo age reversal.
Collapse
Affiliation(s)
- Wayne Mitchell
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 United States
| | - Ludger J.E. Goeminne
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 United States
| | - Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 United States
| | - Sirui Zhang
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 United States
| | - Julie Y. Chen
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 United States
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115 United States
| | - Kerry A. Pierce
- Broad Institute of MIT and Harvard, Cambridge, MA 01241 United States
| | - Angelina H. Choy
- Broad Institute of MIT and Harvard, Cambridge, MA 01241 United States
| | - Clary B. Clish
- Broad Institute of MIT and Harvard, Cambridge, MA 01241 United States
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115 United States
| | - Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 United States
| |
Collapse
|
14
|
Belsky DW, Baccarelli AA. To promote healthy aging, focus on the environment. NATURE AGING 2023; 3:1334-1344. [PMID: 37946045 DOI: 10.1038/s43587-023-00518-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/27/2023] [Indexed: 11/12/2023]
Abstract
To build health equity for an aging world marked by dramatic disparities in healthy lifespan between countries, regions and population groups, research at the intersections of biology, toxicology and the social and behavioral sciences points the way: to promote healthy aging, focus on the environment. In this Perspective, we suggest that ideas and tools from the emerging field of geroscience offer opportunities to advance the environmental science of aging. Specifically, the capacity to measure the pace and progress of biological processes of aging within individuals from relatively young ages makes it possible to study how changing environments can change aging trajectories from early in life, in time to prevent or delay aging-related disease and disability and build aging health equity.
Collapse
Affiliation(s)
- Daniel W Belsky
- Robert N. Butler Columbia Aging Center and Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA.
| |
Collapse
|
15
|
Raffington L, Schneper L, Mallard T, Fisher J, Vinnik L, Hollis-Hansen K, Notterman DA, Tucker-Drob EM, Mitchell C, Harden KP. Salivary Epigenetic Measures of Body Mass Index and Social Determinants of Health Across Childhood and Adolescence. JAMA Pediatr 2023; 177:1047-1054. [PMID: 37669030 PMCID: PMC10481322 DOI: 10.1001/jamapediatrics.2023.3017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/07/2023] [Indexed: 09/06/2023]
Abstract
Importance Children who are socioeconomically disadvantaged are at increased risk for high body mass index (BMI) and multiple diseases in adulthood. The developmental origins of health and disease hypothesis proposes that early life conditions affect later-life health in a manner that is only partially modifiable by later-life experiences. Objective To examine whether epigenetic measures of BMI developed in adults are valid biomarkers of childhood BMI and if they are sensitive to early life social determinants of health. Design, Setting, and Participants This population-based study of over 3200 children and adolescents aged 8 to 18 years included data from 2 demographically diverse US pediatric cohort studies that combine longitudinal and twin study designs. Analyses were conducted from 2021 to 2022. Exposures Socioeconomic status, marginalized groups. Main Outcome and Measure Salivary epigenetic BMI, BMI. Analyses were conducted to validate the use of saliva epigenetic BMI as a potential biomarker of child BMI and to examine associations between epigenetic BMI and social determinants of health. Results Salivary epigenetic BMI was calculated from 2 cohorts: (1) 1183 individuals aged 8 to 18 years (609 female [51%]; mean age, 13.4 years) from the Texas Twin Project and (2) 2020 children (1011 female [50%]) measured at 9 years of age and 15 years of age from the Future of Families and Child Well-Being Study. Salivary epigenetic BMI was associated with children's BMI (r = 0.36; 95% CI, 0.31-0.40 to r = 0.50; 95% CI, 0.42-0.59). Longitudinal analysis found that epigenetic BMI was highly stable across adolescence but remained both a leading and lagging indicator of BMI change. Twin analyses showed that epigenetic BMI captured differences in BMI between monozygotic twins. Moreover, children from more disadvantaged socioeconomic status (b = -0.13 to -0.15 across samples) and marginalized racial and ethnic groups (b = 0.08-0.34 across samples) had higher epigenetic BMI, even when controlling for concurrent BMI, pubertal development, and tobacco exposure. Socioeconomic status at birth relative to concurrent socioeconomic status best predicted epigenetic BMI in childhood and adolescence (b = -0.15; 95% CI, -0.20 to -0.09). Conclusion and Relevance This study demonstrated that epigenetic measures of BMI calculated from pediatric saliva samples were valid biomarkers of childhood BMI and may be associated with early-life social inequalities. The findings are in line with the hypothesis that early-life conditions are especially important factors in epigenetic regulation of later-life health. Research showing that health later in life is linked to early-life conditions has important implications for the development of early-life interventions that could significantly extend healthy life span.
Collapse
Affiliation(s)
- Laurel Raffington
- Max Planck Research Group Biosocial – Biology, Social Disparities, and Development, Max Planck Institute for Human Development, Berlin, Germany
- Population Research Center, The University of Texas at Austin, Austin
| | - Lisa Schneper
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | - Travis Mallard
- Population Research Center, The University of Texas at Austin, Austin
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Jonah Fisher
- Survey Research Center, University of Michigan, Ann Arbor
| | - Liza Vinnik
- Population Research Center, The University of Texas at Austin, Austin
| | | | - Daniel A. Notterman
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | | | - Colter Mitchell
- Survey Research Center, University of Michigan, Ann Arbor
- Population Studies Center, University of Michigan, Ann Arbor
| | - K. Paige Harden
- Population Research Center, The University of Texas at Austin, Austin
| |
Collapse
|
16
|
Kerepesi C, Gladyshev VN. Intersection clock reveals a rejuvenation event during human embryogenesis. Aging Cell 2023; 22:e13922. [PMID: 37786333 PMCID: PMC10577537 DOI: 10.1111/acel.13922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/08/2023] [Accepted: 06/20/2023] [Indexed: 10/04/2023] Open
Abstract
Recent research revealed a rejuvenation event during early development of mice. Here, by examining epigenetic age dynamics of human embryogenesis, we tested whether a similar event exists in humans. For this purpose, we developed an epigenetic clock method, the intersection clock, that utilizes bisulfite sequencing in a way that maximizes the use of informative CpG sites with no missing clock CpG sites in test samples and applied it to human embryo development data. We observed no changes in the predicted epigenetic age between cleavage stage and blastocyst stage embryos; however, a significant decrease was observed between blastocysts and cells representing the epiblast. Additionally, by applying the intersection clock to datasets spanning pre and postimplantation, we found no significant change in the epigenetic age during preimplantation stages; however, the epigenetic age of postimplantation samples was lower compared to the preimplantation stages. We further investigated the epigenetic age of primed (representing early postimplantation) and naïve (representing preimplantation) pluripotent stem cells and observed that in all cases the epigenetic age of primed cells was significantly lower than that of naïve cells. Together, our data suggest that human embryos are rejuvenated during early embryogenesis. Hence, the rejuvenation event is conserved between the mouse and human, and it occurs around the gastrulation stage in both species. Beyond this advance, the intersection clock opens the way for other epigenetic age studies based on human bisulfite sequencing datasets as opposed to methylation arrays.
Collapse
Affiliation(s)
- Csaba Kerepesi
- Brigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- Institute for Computer Science and Control (SZTAKI), Eötvös Loránd Research NetworkBudapestHungary
| | - Vadim N. Gladyshev
- Brigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
17
|
Thomas A, Belsky D, Gu Y. Healthy Lifestyle Behaviors and Biological Aging in the U.S. National Health and Nutrition Examination Surveys 1999-2018. J Gerontol A Biol Sci Med Sci 2023; 78:1535-1542. [PMID: 36896965 PMCID: PMC10460553 DOI: 10.1093/gerona/glad082] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Indexed: 03/11/2023] Open
Abstract
People who have a balanced diet and engage in more physical activity live longer, healthier lives. This study aimed to test the hypothesis that these associations reflect a slowing of biological processes of aging. We analyzed data from 42 625 participants (aged 20-84 years, 51% female participants) from the National Health and Nutrition Examination Surveys (NHANES), 1999-2018. We calculated adherence to a Mediterranean diet (MeDi) and level of leisure time physical activity (LTPA) using standard methods. We measured biological aging by applying the PhenoAge algorithm, developed using clinical and mortality data from NHANES-III (1988-94), to clinical chemistries measured from a blood draw at the time of the survey. We tested the associations of diet and physical activity measures with biological aging, explored synergies between these health behaviors, and tested heterogeneity in their associations across strata of age, sex, and body mass index. Participants who adhered to the MeDi and who did more LTPA had younger biological ages compared with those who had less-healthy lifestyles (high vs low MeDi tertiles: β = 0.14 standard deviation [SD] [95% confidence interval {CI}: -0.18, -0.11]; high vs sedentary LTPA, β = 0.12 SD [-0.15, -0.09]), in models controlled for demographic and socioeconomic characteristics. Healthy diet and regular physical activity were independently associated with lower clinically defined biological aging, regardless of age, sex, and BMI category.
Collapse
Affiliation(s)
- Aline Thomas
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, USA
| | - Daniel W Belsky
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, New York, USA
- Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Yian Gu
- Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, New York, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Gertrude H. Sergievsky Center, and Department of Neurology, Columbia University, New York, New York, USA
| |
Collapse
|
18
|
Gou M, Li J, Yi L, Li H, Ye X, Wang H, Liu L, Sun B, Zhang S, Zhu Z, Liu J, Liu L. Reprogramming of ovarian aging epigenome by resveratrol. PNAS NEXUS 2023; 2:pgac310. [PMID: 36743471 PMCID: PMC9896145 DOI: 10.1093/pnasnexus/pgac310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/23/2022] [Indexed: 12/26/2022]
Abstract
Resveratrol is an antiaging, antioxidant, and anti-inflammatory natural polyphenolic compound. Growing evidence indicates that resveratrol has potential therapeutic effects for improving aging ovarian function. However, the mechanisms underlying prolonged reproductive longevity remain elusive. We found that resveratrol ameliorates ovarian aging transcriptome, some of which are associated with specific changes in methylome. In addition to known aging transcriptome of oocytes and granulosa cells such as decline in oxidoreductase activity, metabolism and mitochondria function, and elevated DNA damage and apoptosis, actin cytoskeleton are notably downregulated with age, and these defects are mostly rescued by resveratrol. Moreover, the aging-associated hypermethylation of actin cytoskeleton is decreased by resveratrol. In contrast, deletion of Tet2, involved in DNA demethylation, abrogates resveratrol-reprogrammed ovarian aging transcriptome. Consistently, Tet2 deficiency results in additional altered pathways as shown by increased mTOR and Wnt signaling, as well as reduced DNA repair and actin cytoskeleton with mouse age. Moreover, genes associated with oxidoreductase activity and oxidation-reduction process were hypermethylated in Tet2-deficient oocytes from middle-age mice treated with resveratrol, indicating that loss of Tet2 abolishes the antioxidant effect of resveratrol. Taking together, our finding provides a comprehensive landscape of transcriptome and epigenetic changes associated with ovarian aging that can be reprogrammed by resveratrol administration, and suggests that aberrantly increased DNA methylation by Tet2 deficiency promotes additional aging epigenome that cannot be effectively restored to younger state by resveratrol.
Collapse
Affiliation(s)
- Mo Gou
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China
| | - Jie Li
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China
| | - Lizhi Yi
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, 100101 Beijing, China
| | - Huiyu Li
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China
| | - Xiaoying Ye
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China
| | - Huasong Wang
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China
| | - Linlin Liu
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China
| | - Baofa Sun
- Department of Zoology, College of Life Science, Nankai University, Tianjin 300071, China
| | - Song Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science, Nankai University, Tianjin 300071, China
| | - Zhengmao Zhu
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China
| | - Jiang Liu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, 100101 Beijing, China
| | - Lin Liu
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China
- Institute of Translational Medicine, Nankai Union Medical Center, Nankai University, Tianjin 300000, China
| |
Collapse
|
19
|
Raffington L, Schneper L, Mallard T, Fisher J, Vinnik L, Hollis-Hansen K, Notterman DA, Tucker-Drob EM, Mitchell C, Harden KP. Measuring the long arm of childhood in real-time: Epigenetic predictors of BMI and social determinants of health across childhood and adolescence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.20.524709. [PMID: 36712110 PMCID: PMC9882281 DOI: 10.1101/2023.01.20.524709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Children who are socioeconomically disadvantaged are at increased risk for high body mass index (BMI) and multiple diseases in adulthood. The developmental origins of health and disease hypothesis proposes that early life conditions affect later-life health in a manner that is only partially modifiable by later-life experiences. Epigenetic mechanisms may regulate the influence of early life conditions on later life health. Recent epigenetic studies of adult blood samples have identified DNA-methylation sites associated with higher BMI and worse health (epigenetic-BMI). Here, we used longitudinal and twin study designs to examine whether epigenetic predictors of BMI developed in adults are valid biomarkers of child BMI and are sensitive to early life social determinants of health. Salivary epigenetic-BMI was calculated from two samples: (1) N=1,183 8-to-19-year-olds (609 female, mean age=13.4) from the Texas Twin Project (TTP), and (2) N=2,020 children (1,011 female) measured at 9 and 15 years from the Future of Families and Child Well-Being Study (FFCWS). We found that salivary epigenetic-BMI is robustly associated with children's BMI (r=0.36 to r=0.50). Longitudinal analysis suggested that epigenetic-BMI is highly stable across adolescence, but remains both a leading and lagging indicator of BMI change. Twin analyses showed that epigenetic-BMI captures differences in BMI between monozygotic twins. Moreover, children from more disadvantaged socioeconomic status (SES) and marginalized race/ethnic groups had higher epigenetic-BMI, even when controlling for concurrent BMI, pubertal development, and tobacco exposure. SES at birth relative to concurrent SES best predicted epigenetic-BMI in childhood and adolescence. We show for the first time that epigenetic predictors of BMI calculated from pediatric saliva samples are valid biomarkers of childhood BMI that are sensitive to social inequalities. Our findings are in line with the hypothesis that early life conditions are especially important factors in epigenetic regulation of later life health. Research showing that health later in life is linked to early life conditions have important implications for the development of early-life interventions that could significantly extend healthy life span.
Collapse
Affiliation(s)
- Laurel Raffington
- Max Planck Institute for Human Development, Max Planck Research Group Biosocial – Biology, Social Disparities, and Development, Lentzeallee 94, 14195 Berlin, Germany
- Population Research Center, The University of Texas at Austin, Austin, TX, USA
| | - Lisa Schneper
- Department of Molecular Biology, Princeton University, Princeton, NJ
| | - Travis Mallard
- Population Research Center, The University of Texas at Austin, Austin, TX, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Jonah Fisher
- Survey Research Center, University of Michigan, Ann Arbor, MI
| | - Liza Vinnik
- Population Research Center, The University of Texas at Austin, Austin, TX, USA
| | | | | | | | - Colter Mitchell
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Population Studies Center, University of Michigan, Ann Arbor, MI
| | - Kathryn P. Harden
- Population Research Center, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
20
|
Abstract
'Age reprogramming' refers to the process by which the molecular and cellular pathways of a cell that are subject to age-related decline are rejuvenated without passage through an embryonic stage. This process differs from the rejuvenation observed in differentiated derivatives of induced pluripotent stem cells, which involves passage through an embryonic stage and loss of cellular identity. Accordingly, the study of age reprogramming can provide an understanding of how ageing can be reversed while retaining cellular identity and the specialised function(s) of a cell, which will be of benefit to regenerative medicine. Here, we highlight recent work that has provided a more nuanced understanding of age reprogramming and point to some open questions in the field that might be explored in the future.
Collapse
Affiliation(s)
- Prim B. Singh
- Department of Medicine, Nazarbayev University School of Medicine, 5/1 Kerei Zhanibek Khandar Street, Astana 010000, Republic of Kazakhstan
| | - Assem Zhakupova
- Department of Medicine, Nazarbayev University School of Medicine, 5/1 Kerei Zhanibek Khandar Street, Astana 010000, Republic of Kazakhstan
| |
Collapse
|
21
|
Avila-Rieger J, Turney IC, Vonk JMJ, Esie P, Seblova D, Weir VR, Belsky DW, Manly JJ. Socioeconomic Status, Biological Aging, and Memory in a Diverse National Sample of Older US Men and Women. Neurology 2022; 99:e2114-e2124. [PMID: 36038275 PMCID: PMC9651454 DOI: 10.1212/wnl.0000000000201032] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/13/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Exposure to socioeconomic disadvantage is associated with early-onset cognitive aging. Biological aging, the progressive loss of system integrity that occurs as we age, is proposed as a modifiable process mediating this health inequality. We examined whether socioeconomic disparities in cognitive aging in mid-to late-life adults is explained by accelerated biological aging similarly across race, ethnicity, and sex/gender. METHODS Data were from a prospective cohort study of the US Health and Retirement Study DNA methylation substudy. Socioeconomic status (SES) was measured from years of education and household wealth at baseline. The extent and pace of biological aging were quantified using 3 DNA methylation measures: PhenoAge, GrimAge, and DunedinPoAm. Cognitive aging was measured from repeated longitudinal assessments of immediate and delayed word recall. Latent growth curve modeling estimated participants' level of memory performance and rate of decline over 2-11 follow-up assessments spanning 2-20 years. Multiple-group models were estimated to assess whether the relationship between SES and memory trajectories was mediated by biological aging across racial-ethnic by sex/gender subgroups. RESULTS Data from a total of 3,997 adults aged 50-100 years were analyzed. Participants with lower SES had a lower memory performance, had a faster decline, and exhibited accelerated biological aging (SES effect size associations [β] ranged from 0.08 to 0.41). Accelerated biological aging was associated with decreased memory performance and faster memory decline (effect size range 0.03-0.23). SES-biological aging associations were the strongest for White men and women and weakest for Latinx women. The relationship between biological aging measures and memory was weaker for Black participants compared with that for White and Latinx people. In mediation analysis, biological aging accounted for 4%-27% of the SES-memory gradient in White participants. There was little evidence of mediation in Black or Latinx participants. DISCUSSION Among a national sample of mid-to late-life adults, DNA methylation measures of biological aging were variably associated with memory trajectories and SES across White, Black, and Latinx mid-to late-life adults. These results challenge the assumption that DNA methylation biomarkers of aging that were developed in primarily White people can equivalently quantify aging processes affecting cognition in Black and Latinx mid-to late-life adults.
Collapse
Affiliation(s)
- Justina Avila-Rieger
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Indira C Turney
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Jet M J Vonk
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Precious Esie
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Dominika Seblova
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Vanessa R Weir
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Daniel W Belsky
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Jennifer J Manly
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY.
| |
Collapse
|
22
|
Avila-Rieger J, Turney IC, Vonk JMJ, Esie P, Seblova D, Weir VR, Belsky DW, Manly JJ. Socioeconomic Status, Biological Aging, and Memory in a Diverse National Sample of Older US Men and Women. Neurology 2022; 99:e2114-e2124. [PMID: 36038275 PMCID: PMC9651454 DOI: 10.1212/wnl.0000000000201032 10.1212/wnl.0000000000201032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/13/2022] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Exposure to socioeconomic disadvantage is associated with early-onset cognitive aging. Biological aging, the progressive loss of system integrity that occurs as we age, is proposed as a modifiable process mediating this health inequality. We examined whether socioeconomic disparities in cognitive aging in mid-to late-life adults is explained by accelerated biological aging similarly across race, ethnicity, and sex/gender. METHODS Data were from a prospective cohort study of the US Health and Retirement Study DNA methylation substudy. Socioeconomic status (SES) was measured from years of education and household wealth at baseline. The extent and pace of biological aging were quantified using 3 DNA methylation measures: PhenoAge, GrimAge, and DunedinPoAm. Cognitive aging was measured from repeated longitudinal assessments of immediate and delayed word recall. Latent growth curve modeling estimated participants' level of memory performance and rate of decline over 2-11 follow-up assessments spanning 2-20 years. Multiple-group models were estimated to assess whether the relationship between SES and memory trajectories was mediated by biological aging across racial-ethnic by sex/gender subgroups. RESULTS Data from a total of 3,997 adults aged 50-100 years were analyzed. Participants with lower SES had a lower memory performance, had a faster decline, and exhibited accelerated biological aging (SES effect size associations [β] ranged from 0.08 to 0.41). Accelerated biological aging was associated with decreased memory performance and faster memory decline (effect size range 0.03-0.23). SES-biological aging associations were the strongest for White men and women and weakest for Latinx women. The relationship between biological aging measures and memory was weaker for Black participants compared with that for White and Latinx people. In mediation analysis, biological aging accounted for 4%-27% of the SES-memory gradient in White participants. There was little evidence of mediation in Black or Latinx participants. DISCUSSION Among a national sample of mid-to late-life adults, DNA methylation measures of biological aging were variably associated with memory trajectories and SES across White, Black, and Latinx mid-to late-life adults. These results challenge the assumption that DNA methylation biomarkers of aging that were developed in primarily White people can equivalently quantify aging processes affecting cognition in Black and Latinx mid-to late-life adults.
Collapse
Affiliation(s)
- Justina Avila-Rieger
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Indira C Turney
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Jet M J Vonk
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Precious Esie
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Dominika Seblova
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Vanessa R Weir
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Daniel W Belsky
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY
| | - Jennifer J Manly
- From the Taub Institute for Research on Alzheimer's Disease and the Aging Brain (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Gertrude H. Sergievsky Center (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Department of Neurology (J.A.-R., I.C.T., J.M.J.V., D.S., V.R.W., J.J.M.), College of Physicians and Surgeons, Columbia University, New York; Julius Center for Health Sciences and Primary Care (J.M.J.V.), Department of Epidemiology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands; Department of Epidemiology (P.E., D.W.B.), Columbia University Mailman School of Public Health, New York; and Butler Columbia Aging Center (D.W.B.), Columbia University Mailman School of Public Health, New York, NY.
| |
Collapse
|
23
|
Dynamic Transcriptional Landscape of Grass Carp (Ctenopharyngodon idella) Reveals Key Transcriptional Features Involved in Fish Development. Int J Mol Sci 2022; 23:ijms231911547. [PMID: 36232849 PMCID: PMC9569805 DOI: 10.3390/ijms231911547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/23/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
A high-quality baseline transcriptome is a valuable resource for developmental research as well as a useful reference for other studies. We gathered 41 samples representing 11 tissues/organs from 22 important developmental time points within 197 days of fertilization of grass carp eggs in order to systematically examine the role of lncRNAs and alternative splicing in fish development. We created a high-quality grass carp baseline transcriptome with a completeness of up to 93.98 percent by combining strand-specific RNA sequencing and single-molecule real-time RNA sequencing technologies, and we obtained temporal expression profiles of 33,055 genes and 77,582 transcripts during development and tissue differentiation. A family of short interspersed elements was preferentially expressed at the early stage of zygotic activation in grass carp, and its possible regulatory components were discovered through analysis. Additionally, after thoroughly analyzing alternative splicing events, we discovered that retained intron (RI) alternative splicing events change significantly in both zygotic activation and tissue differentiation. During zygotic activation, we also revealed the precise regulatory characteristics of the underlying functional RI events.
Collapse
|
24
|
Raffington L, Belsky DW. Integrating DNA Methylation Measures of Biological Aging into Social Determinants of Health Research. Curr Environ Health Rep 2022; 9:196-210. [PMID: 35181865 DOI: 10.1007/s40572-022-00338-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Acceleration of biological processes of aging is hypothesized to drive excess morbidity and mortality in socially disadvantaged populations. DNA methylation measures of biological aging provide tools for testing this hypothesis. RECENT FINDINGS Next-generation DNA methylation measures of biological aging developed to predict mortality risk and physiological decline are more predictive of morbidity and mortality than the original epigenetic clocks developed to predict chronological age. These new measures show consistent evidence of more advanced and faster biological aging in people exposed to socioeconomic disadvantage and may be able to record the emergence of socially determined health inequalities as early as childhood. Next-generation DNA methylation measures of biological aging also indicate race/ethnic disparities in biological aging. More research is needed on these measures in samples of non-Western and non-White populations. New DNA methylation measures of biological aging open opportunities for refining inference about the causes of social disparities in health and devising policies to eliminate them. Further refining measures of biological aging by including more diversity in samples used for measurement development is a critical priority for the field.
Collapse
Affiliation(s)
- Laurel Raffington
- Department of Psychology, University of Texas at Austin, Austin, TX, USA
- Population Research Center, The University of Texas at Austin, Austin, TX, USA
| | - Daniel W Belsky
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 W 168th St. Rm 413, New York, NY, 10032, USA.
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA.
| |
Collapse
|
25
|
Seale K, Horvath S, Teschendorff A, Eynon N, Voisin S. Making sense of the ageing methylome. Nat Rev Genet 2022; 23:585-605. [PMID: 35501397 DOI: 10.1038/s41576-022-00477-6] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2022] [Indexed: 12/22/2022]
Abstract
Over time, the human DNA methylation landscape accrues substantial damage, which has been associated with a broad range of age-related diseases, including cardiovascular disease and cancer. Various age-related DNA methylation changes have been described, including at the level of individual CpGs, such as differential and variable methylation, and at the level of the whole methylome, including entropy and correlation networks. Here, we review these changes in the ageing methylome as well as the statistical tools that can be used to quantify them. We detail the evidence linking DNA methylation to ageing phenotypes and the longevity strategies aimed at altering both DNA methylation patterns and machinery to extend healthspan and lifespan. Lastly, we discuss theories on the mechanistic causes of epigenetic ageing.
Collapse
Affiliation(s)
- Kirsten Seale
- Institute for Health and Sport (iHeS), Victoria University, Footscray, Melbourne, Victoria, Australia
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Altos Labs, San Diego, CA, USA
| | - Andrew Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China.,UCL Cancer Institute, University College London, London, UK
| | - Nir Eynon
- Institute for Health and Sport (iHeS), Victoria University, Footscray, Melbourne, Victoria, Australia.
| | - Sarah Voisin
- Institute for Health and Sport (iHeS), Victoria University, Footscray, Melbourne, Victoria, Australia.
| |
Collapse
|
26
|
McGill MG, Pokhvisneva I, Clappison AS, McEwen LM, Beijers R, Tollenaar MS, Pham H, Kee MZL, Garg E, de Mendonça Filho EJ, Karnani N, Silveira PP, Kobor MS, de Weerth C, Meaney MJ, O'Donnell KJ. Maternal Prenatal Anxiety and the Fetal Origins of Epigenetic Aging. Biol Psychiatry 2022; 91:303-312. [PMID: 34756561 DOI: 10.1016/j.biopsych.2021.07.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND The fetal origins of mental health is a well-established framework that currently lacks a robust index of the biological embedding of prenatal adversity. The Pediatric-Buccal-Epigenetic (PedBE) clock is a novel epigenetic tool that associates with aspects of the prenatal environment, but additional validation in longitudinal datasets is required. Likewise, the relationship between prenatal maternal mental health and the PedBE clock has not been described. METHODS Longitudinal cohorts from the Netherlands (Basal Influences on Baby Development [BIBO] n = 165) and Singapore (Growing Up in Singapore Towards Healthy Outcomes [GUSTO] n = 340) provided data on prenatal maternal anxiety and longitudinal assessments of buccal cell-derived genome-wide DNA methylation assessed at 6 and 10 years of age in BIBO, and at 3, 9, and 48 months of age in GUSTO. Measures of epigenetic age acceleration were calculated using the PedBE clock and benchmarked against an established multi-tissue epigenetic predictor. RESULTS Prenatal maternal anxiety predicted child PedBE epigenetic age acceleration in both cohorts, with effects largely restricted to males and not females. These results were independent of obstetric, socioeconomic, and genetic risk factors, with a larger effect size for prenatal anxiety than depression. PedBE age acceleration predicted increased externalizing symptoms in males from mid- to late childhood in the BIBO cohort only. CONCLUSIONS These findings point to the fetal origins of epigenetic age acceleration and reveal an increased sensitivity in males. Convergent evidence underscores the societal importance of providing timely and effective mental health support to pregnant individuals, which may have lasting consequences for both mother and child.
Collapse
Affiliation(s)
- Megan G McGill
- Douglas Hospital Research Centre, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, and Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, Quebec, Canada
| | - Irina Pokhvisneva
- Douglas Hospital Research Centre, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, and Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, Quebec, Canada
| | - Andrew S Clappison
- Douglas Hospital Research Centre, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, and Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, Quebec, Canada
| | - Lisa M McEwen
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Roseriet Beijers
- Department of Developmental Psychology, Radboud University, Nijmegen, the Netherlands; Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, the Netherlands
| | - Marieke S Tollenaar
- Department of Clinical Psychology, Institute of Psychology and Leiden Institute for Brain and Cognition, Leiden University, Leiden, the Netherlands
| | - Hung Pham
- Yale Child Study Center and Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut
| | | | - Elika Garg
- Douglas Hospital Research Centre, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, and Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, Quebec, Canada
| | | | - Neerja Karnani
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore
| | - Patricia P Silveira
- Douglas Hospital Research Centre, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, and Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, Quebec, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada; Child and Brain Development Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada
| | - Carolina de Weerth
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboudumc, Nijmegen, the Netherlands
| | - Michael J Meaney
- Douglas Hospital Research Centre, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, and Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, Quebec, Canada; Singapore Institute for Clinical Sciences, Singapore; Child and Brain Development Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kieran J O'Donnell
- Douglas Hospital Research Centre, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, and Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, Quebec, Canada; Yale Child Study Center and Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut; Child and Brain Development Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada.
| |
Collapse
|
27
|
Meron E, Thaysen M, Angeli S, Antebi A, Barzilai N, Baur JA, Bekker-Jensen S, Birkisdottir M, Bischof E, Bruening J, Brunet A, Buchwalter A, Cabreiro F, Cai S, Chen BH, Ermolaeva M, Ewald CY, Ferrucci L, Florian MC, Fortney K, Freund A, Georgievskaya A, Gladyshev VN, Glass D, Golato T, Gorbunova V, Hoejimakers J, Houtkooper RH, Jager S, Jaksch F, Janssens G, Jensen MB, Kaeberlein M, Karsenty G, de Keizer P, Kennedy B, Kirkland JL, Kjaer M, Kroemer G, Lee KF, Lemaitre JM, Liaskos D, Longo VD, Lu YX, MacArthur MR, Maier AB, Manakanatas C, Mitchell SJ, Moskalev A, Niedernhofer L, Ozerov I, Partridge L, Passegué E, Petr MA, Peyer J, Radenkovic D, Rando TA, Rattan S, Riedel CG, Rudolph L, Ai R, Serrano M, Schumacher B, Sinclair DA, Smith R, Suh Y, Taub P, Trapp A, Trendelenburg AU, Valenzano DR, Verburgh K, Verdin E, Vijg J, Westendorp RGJ, Zonari A, Bakula D, Zhavoronkov A, Scheibye-Knudsen M. Meeting Report: Aging Research and Drug Discovery. Aging (Albany NY) 2022. [PMID: 35089871 PMCID: PMC8833115 DOI: 10.18632/aging.203859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Aging is the single largest risk factor for most chronic diseases, and thus possesses large socioeconomic interest to continuously aging societies. Consequently, the field of aging research is expanding alongside a growing focus from the industry and investors in aging research. This year’s 8th Annual Aging Research and Drug Discovery (ARDD) meeting was organized as a hybrid meeting from August 30th to September 3rd 2021 with more than 130 attendees participating on-site at the Ceremonial Hall at University of Copenhagen, Denmark, and 1800 engaging online. The conference comprised of presentations from 75 speakers focusing on new research in topics including mechanisms of aging and how these can be modulated as well as the use of AI and new standards of practices within aging research. This year, a longevity workshop was included to build stronger connections with the clinical community.
Collapse
Affiliation(s)
- Esther Meron
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Maria Thaysen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Suzanne Angeli
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Adam Antebi
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Nir Barzilai
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Institute for Aging Research, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Joseph A Baur
- Smilow Center for Translational Research, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Simon Bekker-Jensen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Maria Birkisdottir
- Department of Molecular Genetics, Erasmus MC, Rotterdam, Netherlands.,Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Evelyne Bischof
- Shanghai University of Medicine and Health Sciences, College of Clinical Medicine, Shanghai, China
| | - Jens Bruening
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Anne Brunet
- Department of Genetics, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Abigail Buchwalter
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Filipe Cabreiro
- Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK.,CECAD Research Center, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Shiqing Cai
- Institute of Neuroscience, Chinese Academy of Science, Shanghai, China
| | - Brian H Chen
- FOXO Technologies Inc, Minneapolis, MN 55402, USA.,The Herbert Wertheim School of Public Health and Human Longevity Science, UC San Diego, La Jolla, CA 92093, USA
| | | | - Collin Y Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach CH-8603, Switzerland
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | | | | - Adam Freund
- Arda Therapeutics, San Carlos, CA 94070, USA
| | | | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David Glass
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY 14627, USA
| | - Jan Hoejimakers
- Department of Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Sibylle Jager
- L'Oréal Research and Innovation, Aulnay-sous-Bois, France
| | | | - Georges Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Matt Kaeberlein
- Departments of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Gerard Karsenty
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Peter de Keizer
- Department of Molecular Cancer Research, Center for Molecular Medicine, Division of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Brian Kennedy
- Buck Institute for Research on Aging, Novato, CA 94945, USA.,Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University Singapore, Singapore.,Center for Healthy Longevity, National University Health System, Singapore
| | - James L Kirkland
- Division of General Internal Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Michael Kjaer
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, Inserm U1138, Paris, France
| | - Kai-Fu Lee
- Sinovation Ventures and Sinovation AI Institute, Beijing, China
| | - Jean-Marc Lemaitre
- Institute for Regenerative Medicine and Biotherapies, INSERM UMR 1183, Montpellier, France
| | | | - Valter D Longo
- USC Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Yu-Xuan Lu
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Michael R MacArthur
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Andrea B Maier
- Center for Healthy Longevity, National University Health System, Singapore.,Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | | | - Sarah J Mitchell
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Alexey Moskalev
- Institute of Biology of FRC Komi Science Center of Ural Division of RAS, Syktyvkar, Russia.,Russian Clinical and Research Center of Gerontology, Moscow, Russia
| | - Laura Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ivan Ozerov
- Insilico Medicine, Hong Kong Science and Technology Park, Hong Kong
| | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Michael A Petr
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.,Tracked.bio, Copenhagen, Denmark
| | | | - Dina Radenkovic
- Hooke London by Health and Longevity Optimisation, London, UK
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences and Paul F. Glenn Center for Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Suresh Rattan
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Christian G Riedel
- Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | | | - Ruixue Ai
- Department of Clinical Molecular Biology
- UiO, University of Oslo and Akershus University Hospital, Norway
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Björn Schumacher
- CECAD Research Center, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - David A Sinclair
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, MA 94107, USA
| | | | - Yousin Suh
- Departments of Obstetrics and Gynecology, Genetics and Development, Columbia University, New York, NY 10027, USA
| | - Pam Taub
- Division of Cardiovascular Medicine, University of California, San Diego, CA 92093, USA
| | - Alexandre Trapp
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Dario Riccardo Valenzano
- Max Planck Institute for Biology of Ageing, Cologne, Germany.,Leibniz Institute on Aging, Jena, Germany
| | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Jan Vijg
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | - Daniela Bakula
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Alex Zhavoronkov
- Insilico Medicine, Hong Kong Science and Technology Park, Hong Kong
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Zhang B, Trapp A, Kerepesi C, Gladyshev VN. Emerging rejuvenation strategies-Reducing the biological age. Aging Cell 2022; 21:e13538. [PMID: 34972247 PMCID: PMC8761015 DOI: 10.1111/acel.13538] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/02/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022] Open
Abstract
Several interventions have recently emerged that were proposed to reverse rather than just attenuate aging, but the criteria for what it takes to achieve rejuvenation remain controversial. Distinguishing potential rejuvenation therapies from other longevity interventions, such as those that slow down aging, is challenging, and these anti-aging strategies are often referred to interchangeably. We suggest that the prerequisite for a rejuvenation intervention is a robust, sustained, and systemic reduction in biological age, which can be assessed by biomarkers of aging, such as epigenetic clocks. We discuss known and putative rejuvenation interventions and comparatively analyze them to explore underlying mechanisms.
Collapse
Affiliation(s)
- Bohan Zhang
- Division of GeneticsDepartment of MedicineHarvard Medical SchoolBrigham and Women’s HospitalBostonMassachusettsUSA
| | - Alexandre Trapp
- Division of GeneticsDepartment of MedicineHarvard Medical SchoolBrigham and Women’s HospitalBostonMassachusettsUSA
| | - Csaba Kerepesi
- Division of GeneticsDepartment of MedicineHarvard Medical SchoolBrigham and Women’s HospitalBostonMassachusettsUSA
| | - Vadim N. Gladyshev
- Division of GeneticsDepartment of MedicineHarvard Medical SchoolBrigham and Women’s HospitalBostonMassachusettsUSA
| |
Collapse
|
29
|
Kasselimi E, Pefani DE, Taraviras S, Lygerou Z. Ribosomal DNA and the nucleolus at the heart of aging. Trends Biochem Sci 2022; 47:328-341. [DOI: 10.1016/j.tibs.2021.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/15/2022]
|
30
|
Affiliation(s)
- K Lenhard Rudolph
- Research Group on Stem Cell and Metabolism Aging, Leibniz Institute on Aging and Fritz Lipmann Institute (FLI), University Hospital Jena (UKJ), Friedrich Schiller University, Jena, Germany.
| |
Collapse
|
31
|
Abstract
DNA methylation dynamics emerged as a promising biomarker of mammalian aging, with multivariate machine learning models ('epigenetic clocks') enabling measurement of biological age in bulk tissue samples. However, intrinsically sparse and binarized methylation profiles of individual cells have so far precluded the assessment of aging in single-cell data. Here, we introduce scAge, a statistical framework for epigenetic age profiling at single-cell resolution, and validate our approach in mice. Our method recapitulates the chronological age of tissues, while uncovering heterogeneity among cells. We show accurate tracking of the aging process in hepatocytes, demonstrate attenuated epigenetic aging in muscle stem cells, and track age dynamics in embryonic stem cells. We also use scAge to reveal, at the single-cell level, a natural and stratified rejuvenation event occurring during early embryogenesis. We provide our framework as a resource to enable exploration of epigenetic aging trajectories at single-cell resolution.
Collapse
|
32
|
A toolkit for quantification of biological age from blood chemistry and organ function test data: BioAge. GeroScience 2021; 43:2795-2808. [PMID: 34725754 DOI: 10.1007/s11357-021-00480-5] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/22/2021] [Indexed: 10/19/2022] Open
Abstract
Methods to quantify biological aging are emerging as new measurement tools for epidemiology and population science and have been proposed as surrogate measures for healthy lifespan extension in geroscience clinical trials. Publicly available software packages to compute biological aging measurements from DNA methylation data have accelerated dissemination of these measures and generated rapid gains in knowledge about how different measures perform in a range of datasets. Biological age measures derived from blood chemistry data were introduced at the same time as the DNA methylation measures and, in multiple studies, demonstrate superior performance to these measures in prediction of healthy lifespan. However, their dissemination has been slow by comparison, resulting in a significant gap in knowledge. We developed a software package to help address this knowledge gap. The BioAge R package, available for download at GitHub ( http://github.com/dayoonkwon/BioAge ), implements three published methods to quantify biological aging based on analysis of chronological age and mortality risk: Klemera-Doubal biological age, PhenoAge, and homeostatic dysregulation. The package allows users to parametrize measurement algorithms using custom sets of biomarkers, to compare the resulting measurements to published versions of the Klemera-Doubal method and PhenoAge algorithms, and to score the measurements in new datasets. We applied BioAge to safety lab data from the CALERIE™ randomized controlled trial, the first-ever human trial of long-term calorie restriction in healthy, non-obese adults, to test effects of intervention on biological aging. Results contribute evidence that CALERIE intervention slowed biological aging. BioAge is a toolkit to facilitate measurement of biological age for geroscience.
Collapse
|
33
|
Searching for female reproductive aging and longevity biomarkers. Aging (Albany NY) 2021; 13:16873-16894. [PMID: 34156973 PMCID: PMC8266318 DOI: 10.18632/aging.203206] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 05/31/2021] [Indexed: 12/21/2022]
Abstract
Female reproductive aging is, in a way, a biological phenomenon that develops along canonical molecular pathways; however, it has particular features. Recent studies revealed complexity of the interconnections between reproductive aging and aging of other systems, and even suggested a cause-effect uncertainty between them. It was also shown that reproductive aging can impact aging processes in an organism at the level of cells, tissues, organs, and systems. Women at the end of their reproductive lives are characterized by the accelerated incidence of age-related diseases. Timing of the onset of menarche and menopause and variability in the duration of reproductive life carry a latent social risk: not having enough information about the reproductive potential, women keep on postponing childbirth. Identification and use of the most accurate and sensitive aging biomarkers enable the prediction of menopause timing and quantification of the true biological and reproductive ages of an organism. We discuss current views on reproductive aging and peculiarities of using available biomarkers of aging. We also consider latest advances in the search for potential genetic markers of reproductive aging. Finally, we posit the importance of determining the female biological age and highlight potential research directions in this area.
Collapse
|
34
|
Kerepesi C, Zhang B, Lee SG, Trapp A, Gladyshev VN. Epigenetic clocks reveal a rejuvenation event during embryogenesis followed by aging. SCIENCE ADVANCES 2021; 7:eabg6082. [PMID: 34172448 PMCID: PMC8232908 DOI: 10.1126/sciadv.abg6082] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 05/12/2021] [Indexed: 05/05/2023]
Abstract
The notion that the germ line does not age goes back to the 19th-century ideas of August Weismann. However, being metabolically active, the germ line accumulates damage and other changes over time, i.e., it ages. For new life to begin in the same young state, the germ line must be rejuvenated in the offspring. Here, we developed a multi-tissue epigenetic clock and applied it, together with other aging clocks, to track changes in biological age during mouse and human prenatal development. This analysis revealed a significant decrease in biological age, i.e., rejuvenation, during early stages of embryogenesis, followed by an increase in later stages. We further found that pluripotent stem cells do not age even after extensive passaging and that the examined epigenetic age dynamics is conserved across species. Overall, this study uncovers a natural rejuvenation event during embryogenesis and suggests that the minimal biological age (ground zero) marks the beginning of organismal aging.
Collapse
Affiliation(s)
- Csaba Kerepesi
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Bohan Zhang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Sang-Goo Lee
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Alexandre Trapp
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
35
|
Raffington L, Belsky DW, Kothari M, Malanchini M, Tucker-Drob EM, Harden KP. Socioeconomic Disadvantage and the Pace of Biological Aging in Children. Pediatrics 2021; 147:e2020024406. [PMID: 34001641 PMCID: PMC8785753 DOI: 10.1542/peds.2020-024406] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Children who grow up in socioeconomic disadvantage face increased burden of disease and disability throughout their lives. One hypothesized mechanism for this increased burden is that early-life disadvantage accelerates biological processes of aging, increasing vulnerability to subsequent disease. To evaluate this hypothesis and the potential impact of preventive interventions, measures are needed that can quantify early acceleration of biological aging in childhood. METHODS Saliva DNA methylation and socioeconomic circumstances were measured in N = 600 children and adolescents aged 8 to 18 years (48% female) participating in the Texas Twin Project. We measured pace of biological aging using the DunedinPoAm DNA methylation algorithm, developed to quantify the pace-of-aging-related decline in system integrity. We tested if children in more disadvantaged families and neighborhoods exhibited a faster pace of aging as compared with children in more affluent contexts. RESULTS Children living in more disadvantaged families and neighborhoods exhibited a faster DunedinPoAm-measured pace of aging (r = 0.18; P = .001 for both). Latinx-identifying children exhibited a faster DunedinPoAm-measured pace of aging compared with both White- and Latinx White-identifying children, consistent with higher levels of disadvantage in this group. Children with more advanced pubertal development, higher BMI, and more tobacco exposure exhibited faster a faster DunedinPoAm-measured pace of aging. However, DunedinPoAm-measured pace of aging associations with socioeconomic disadvantage were robust to control for these factors. CONCLUSIONS Children growing up under conditions of socioeconomic disadvantage exhibit a faster pace of biological aging. DNA methylation pace of aging might be useful as a surrogate end point in evaluation of programs and policies to address the childhood social determinants of lifelong health disparities.
Collapse
Affiliation(s)
- Laurel Raffington
- Department of Psychology and
- Population Research Center, The University of Texas at Austin, Austin, Texas
| | - Daniel W Belsky
- Department of Epidemiology and
- The Robert N. Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, New York; and
| | - Meeraj Kothari
- The Robert N. Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, New York; and
| | - Margherita Malanchini
- Department of Psychology and
- Population Research Center, The University of Texas at Austin, Austin, Texas
- Department of Biological and Experimental Psychology, Queen Mary University of London, London, United Kingdom
| | - Elliot M Tucker-Drob
- Department of Psychology and
- Population Research Center, The University of Texas at Austin, Austin, Texas
- Contributed equally as co-lead authors
| | - K Paige Harden
- Department of Psychology and
- Population Research Center, The University of Texas at Austin, Austin, Texas
- Contributed equally as co-lead authors
| |
Collapse
|
36
|
Abstract
Aging has largely been defined by analog measures of organ and organismal dysfunction. This has led to the characterization of aging processes at the molecular and cellular levels that underlie these gradual changes. However, current knowledge does not fully explain the growing body of data emerging from large epidemiological, systems biology, and single cell studies of entire organisms pointing to varied rates of aging between individuals (different functionality and lifespan), across lifespan (asynchronous aging), and within an organism at the tissue and organ levels (aging mosaicism). Here we consider these inhomogeneities in the broader context of the rate of aging and from the perspective of underlying cellular changes. These changes reflect genetic, environmental, and stochastic factors that cells integrate to adopt new homeostatic, albeit less functional, states, such as cellular senescence. In this sense, cellular aging can be viewed, at least in part, as a quantal process we refer to as "digital aging". Nevertheless, analog declines of tissue dysfunction and organ failure with age could be the sum of underlying digital events. Importantly, cellular aging, digital or otherwise, is not uniform across time or space within the organism or between organisms of the same species. Certain tissues may exhibit earliest signs of cellular aging, acting as drivers for organismal aging as signals from those driver cells within those tissues may accelerate the aging of other cells locally or even systemically. Advanced methodologies at the systems level and at the single cell level are likely to continue to refine our understanding to the processes of how cells and tissues age and how the integration of those processes leads to the complexities of individual, organismal aging.
Collapse
|